BACKGROUND OF THE INVENTION
[0001] Colorectal cancer (CRC) is the third most common cancer and the third leading cause
of cancer mortality worldwide, with an estimated incidence of over 1 million new cases
and a mortality of more than 500,000 deaths per year. Several intrinsic (
e.g., age, male gender, ethnicity, diabetes mellitus, obesity and inflammatory bowel disease)
and extrinsic (
e.g., cigarette smoking, inadequate intake of fiber, high consumption of alcohol, red meat
and high-fat diet) factors are associated with increased risks for colorectal cancer.
The epidemiology of colon cancer is under dynamic changes owing to the changing prevalence
and distribution of risk factors. In this regard, colon cancer incidence in many developing
countries, including Asian countries, has increased 2- to 4-fold over the last two
decades and has now reached an alarming rate, with Westernization of diet playing
a pivotal role.
[0002] About 38 trillion bacteria exist in the human intestine. In view of their symbiotic
and co-operative relationship with the human body, these bacteria have a close association
with the pathogenesis and progression of colon cancer. The association of colon cancer
with altered gut microbiota has been studied in different populations, with certain
bacterial species identified for their potential roles, either beneficial or detrimental,
in tumourigenesis. Disease management and facilitation of treatment among colon cancer
patients by way of modifying the profile of gut microorganisms is a highly desirable
means of medical intervention due to its high efficacy, low cost, and low risk of
side effects. Immune checkpoint blockade (ICB) therapy has shown great promise in
cancer treatment and has seen increased use in colorectal cancer treatment in the
past decade. Also known as anti-PD1/PDL1 (programmed cell death protein 1/programmed
death ligand 1) and/or anti-CTLA4 (cytotoxic T-lymphocyte-associated antigen-4) therapy,
ICB therapy acts by removing the "brake" signal of T cell activation and elicits an
anti-cancer immune response against cancer cells in order to suppress disease progression.
There are notable limitations associated with this therapeutic approach, however,
including drug resistance. Thus, new methods and compositions for the purposes of
improving colon cancer treatment efficacy are desired.
BRIEF SUMMARY OF THE INVENTION
[0003] The inventors discovered that certain gut microbial species and their metabolites
can suppress the proliferation of colorectal cancer cells, especially when applied
in combination with immunotherapeutic agents for treating colon cancer. The identified
microorganisms and metabolites provide new methods and compositions for enhancing
or improving the therapeutic efficacy of immunotherapy for the treatment of colorectal
cancer.
[0004] In a first aspect, the present invention provides a composition for use in improving
or enhancing immunotherapy efficacy in a human patient being treated for colon cancer.
The composition contains an effective amount of (1) any one or any two or three of
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12; and (2) one or more physiologically acceptable excipients. In some embodiments,
the composition comprises an effective amount of two or three of
L. gallinarum, R. intestinalis, and
S. salivarius K12. In some embodiments, the composition comprises a total of about 10
6 to about 10
14 colony-forming unit (CFU) of any one or any two or all three of
L. gallinarum, R. intestinalis, and
S.
salivarius K12. In some embodiments, the composition further comprises indole-3-carboxylic acid (ICA).
In some embodiments, the composition further comprises butyrate. In some embodiments,
the composition is formulated for oral ingestion,
e.g., in the form of a food or beverage item. In some embodiments, the composition is formulated
in a daily dosage comprising about 5 × 10
9 CFU of
L. gallinarum per kg patient bodyweight. In some embodiments, the composition is formulated in
a daily dosage comprising about 5 × 10
9 CFU of
R. intestinalis per kg recipient bodyweight. In some embodiments, the composition is formulated in
a daily dosage comprising about 5 × 10
9 CFU of
S.
salivarius K12 per kg recipient bodyweight.
[0005] In the second aspect, the present invention provides a method for improving or enhancing
or augmenting immunotherapy efficacy in the treatment of colon cancer by administering
to a subject in need thereof an effective amount of the composition described above
and herein, namely containing an effective amount of (1) any one or more of
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12; and (2) one or more physiologically acceptable excipients. In some embodiments,
the immunotherapy comprises administration of an effective amount of a PD1 inhibitor
or a PDL1 inhibitor. In some embodiments, the PD1 or PDL1 inhibitor is an anti-PD1
or an-PDL1 antibody, respectively. In some embodiments, the PD1 inhibitor or PDL1
inhibitor is administered via injection,
e.g., via intravenous infusion. In some embodiments, the claimed method includes a step
of administering to the subject one composition comprising any one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12. In some embodiments, the method comprises the step of administering to the subject
two or more compositions comprising any two or all three of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally further comprising ICA and/or butyrate. In some embodiments, the administering
step comprises oral ingestion of the composition, for example, the administering step
comprises oral ingestion prior to or with food intake.
[0006] In a related aspect, the present invention provides a novel use of a composition
for improving or enhancing immunotherapy efficacy in the treatment of colorectal cancer.
The composition contains an effective amount of (1) any one or more of
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12; and (2) one or more physiologically acceptable excipients. In some embodiments,
the immunotherapy comprises administration of an effective amount of a PD1 inhibitor
or a PDL1 inhibitor. In some embodiments, the PD1/PDL1 inhibitor is an anti-PD1/PDL-1
antibody, respectively. In some embodiments, the PD1 inhibitor or PDL1 inhibitor is
administered via injection,
e.g., via intravenous infusion. In some embodiments, the claimed method includes a step
of administering to the subject one composition comprising any one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12. In some embodiments, the method comprises the step of administering to the subject
two or more compositions comprising any two or all three of
L. gallinarum, R. intestinalis, and
S.
salivarius K12, optionally further comprising ICA and/or butyrate. In some embodiments, the administering
step comprises oral ingestion of the composition, for example, the administering step
comprises oral ingestion prior to or with food intake.
[0007] In a third aspect, the present invention provides a kit for improving or enhancing
immunotherapy efficacy in the treatment of colorectal cancer. The kit comprises a
plurality of compositions each comprising an effective amount of one or more of
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12, optionally further comprising an effective amount of ICA and/or butyrate. In
some embodiments, the kit includes at least two compositions, the first comprising
an effective amount of any one or two or three of
L. gallinarum, R. intestinalis, and
S. salivarius K12, and the second comprising an effective amount of a PD-1 inhibitor or a PD-L1 inhibitor,
e.g., the PD1/PDL1 inhibitor is an anti-PD1/PD-L1 antibody, respectively. In some embodiments,
the first composition is formulated for oral ingestion. In some embodiments, the second
composition is formulated for injection. In some embodiments, the first composition
is formulated in a daily dosage comprising about 10
8 CFU of any one of
L. gallinarum, R. intestinalis, or
S. salivarius K12.
BRIEF DESCRIPTION OF THE DRAWINGS
[0008]
Figures 1A - 1G illustrate L. gallinarum improved anti-PD1 efficacy in MC38 and CT26 syngeneic mouse models. Figure 1A is a schematic diagram of experimental design for syngeneic mouse model.
L. gallinarum, in combination with anti-PD1 therapy, significantly inhibited tumour growth in MC38
syngeneic mouse model (MSI-H model), as evidenced by representative tumour pictures
as illustrated in Figure 1B, tumour weight as illustrated in Figure 1C, and tumour
volume as illustrated in Figure 1D. L. gallinarum also promoted anti-PD1 efficacy in CT26 syngeneic mouse model, an MSI-L model, as
supported by representative tumour pictures as illustrated in Figure 1E, tumour weight
as illustrated in Figure 1F, and tumour volume as illustrated in Figure 1G. **P<0.01,
****P<0.0001.
Figures 2A - 2F illustrate L. gallinarum reduced Foxp3+ CD25+ Treg infiltration and increased IFNγ+ CD8+ T cells in the tumour
microenvironment. Figure 2A shows representative flow cytometry plots of Foxp3+ CD25+ Treg. L. gallinarum significantly reduced Foxp3+ CD25+ Treg infiltration in MC38 tumours as illustrated
in Figure 2B, and CT26 tumours as illustrated in Figure 2C. Figure 2D shows representative
flow cytometry histogram of IFNγ+ CD8+ T cells. L. gallinarum, in combination with anti-PD1, increased IFNγ+ CD8+ T cells in MC38 tumours as illustrated
in Figure 2E, and CT26 tumours as illustrated in Figure 2F. *P<0.05, **P<0.01, ***P<0.001.
Figures 3A - 3H show L. gallinarum improved anti-PD1 efficacy in AOM/DSS-induced CRC mouse model. Figure 3A is a schematic diagram of experimental design for AOM/DSS-induced CRC model.
Figure 3B shows representative colonoscopy images, and Figure 3C shows representative
colon images of AOM/DSS-induced CRC tumourigenesis mouse model. L. gallinarum, in combination with anti-PD1, reduced tumour number as illustrated in Figure 3D,
tumour load as illustrated in Figure 3E, and the number of large tumours (diameter
larger than 2mm) as illustrated in Figure 3F. Figure 3G illustrates the percentage
of Foxp3+ CD25+ Tregs in colonic tumour tissues. Figure 3H shows the percentage of
IFNγ+ CD8+ T cells in colonic tumour tissues. *P<0.05, **P<0.01, ***P<0.001.
Figures 4A - 4E show L. gallinarum produced tryptophan metabolites in vitro and in vivo. Figure 4A is a heatmap analysis of BHI, E. coli and L. gallinarum culture supernatant in vitro. Figure 4B is a heatmap analysis of stool samples from MC38 syngeneic mouse model
that revealed a differential abundance of metabolites in BHI-, E. coli- and L. gallinarum-treated mice. Figure 4C shows Indole-3-carboxaldehyde (lAld) was enriched in stool
samples of L. gallinarum-treated mice in MC38 syngeneic mouse model. No significant difference of indole-3-carboxaldehyde
(lAld) was observed in serum samples between groups as shown in Figure 4D. Figure
4E shows Indole-3-carboxylic acid (ICA) was enriched in serum of L. gallinarum-treated mice in MC38 syngeneic mouse model. *P<0.05, **P<0.01, ****P<0.0001.
Figures 5A - 5H illustrate L. gallinarum and ICA inhibited IDO1 expression and Kyn production in tumour. L. gallinarum reduced serum kynurenine level (Figure 5A), tumour kynurenine level (Figure 5B),
kynurenine-to-tryptophan ratio in tumours in MC38 syngeneic mouse model (Figure 5C).
RNA sequencing revealed a differential gene expression between BHI plus anti-PD1 group
versus L. gallinarum plus anti-PD1 group as illustrated in Figure 5D. Figure 5E shows the enrichment plot
of tryptophan metabolism pathway. Figure 5F illustrates IDO1 mRNA expression of tumour
tissues from MC38 and CT26 syngeneic mouse model. Figure 5G shows immunohistochemical
staining of IDO1 in tumour tissues of CT26 syngeneic mouse model. Figure 5H shows
the effect of ICA (5µM) on IDO1 mRNA expression in HCT116 and LoVo cell lines. *P<0.05,
**P<0.01, ***P<0.001, ****P<0.0001.
Figures 6A - 6G show ICA outcompeted Kyn and inhibited Kyn-mediated AHR activation on CD4+ T cells. Effect of L. gallinarum conditioned medium (Figure 6A), indole-3-carboxaldhye (lAld) (Figure 6B), and indole-3-carboxylic
acid (ICA) on Foxp3+ Treg differentiation (Figure 6C). Figure 6D illustrates the effect
of ICA and Kyn (50µM) on Foxp3+ Treg differentiation. ICA antagonized Kyn-mediated
Treg differentiation in a dose-dependent manner. Figure 6E shows CYP1B1 expression
of CD4+ T cells treated with different dose of ICA and Kyn (50µM). Figure 6F shows
CH-233191 (AHR antagonist) (10µM) abolished the effect of ICA and Kyn. Figure 6G is
a surface plasmon resonance (SPR) assay of ICA and Kyn on human AHR protein. ***P<0.001,
****P<0.0001.
Figures 7A- 7H show ICA improved anti-PD1 efficacy and was reversed by Kyn supplementation. ICA in combination with anti-PD1 impeded tumour growth, as evidenced by representative
tumour pictures as illustrated in Figure 7A, tumour weight as illustrated in Figure
7B, and tumour volume as illustrated in Figure 7C, in CT26 syngeneic mouse model.
Figure 7D shows the percentage of Foxp3+ CD25+ Treg in tumour tissues. Figure 7E shows
the percentage of IFNγ+ CD8+ T cells of tumour tissues. Figure 7F shows Kynurenine
level in serum, Figure 7G shows Kynurenine level in tumour tissues, and Figure 7H
shows Kynurenine-to-tryptophan ratio in tumour tissues. *P<0.05, **P<0.01, ***P<0.001.
Figures 8A - 8D show R. intestinalis and butyric acid are depleted in faecal samples of CRC patients. Arcsine square root transferred relative abundance of R. intestinalis in published metagenomic cohorts (Figure 8A) and in-house metagenomic cohort (Figure
8B) of colorectal cancer patients and healthy controls. CN, China; DE, Germany; FR,
France; JP, Japan. Figure 8C is volcano plots of significantly altered human faecal
metabolites between CRC and NC. Enriched metabolites are labelled as red dots and
depleted ones are labelled as blue. -Logic P (0.05) was indicated with the horizontal
dash line. CRC, colorectal cancer. NC, normal control. Figure 8D is a graph showing
the relative abundance of R. intestinalis in a France cohort of responders and non-responders to immune checkpoint inhibitors.
Figures 9A - 9K show R. intestinalis reduces colorectal carcinogenesis in ApcMin/+ and AOM-induced mice. Figure 9A is schematic diagram showing the experimental design and timeline of ApcMin/+ mice. Figure 9B shows representative colonic morphologies (left panel) and colonoscopy
video captures (right panel) of ApcMin/+ mice. Colon tumour incidence (Figure 9C), colon and small intestine tumour number
and tumour load (total tumour area, mm2) (Figure 9D) of ApcMin/+ mice. Figure 9E are representative H & E staining images (scale bar=200µm) and Ki-67
IHC staining (scale bar=50µm) of ApcMin/+ mice. Figure 9F is a schematic diagram showing the experimental design and timeline
of AOM-induced mice. Figure 9G are representative colonoscopy video captures (upper
panel) and incidence of AOM-induced mice with colonoscopy observable tumour (down
panel). Figure 9H shows representative colonic morphologies of AOM-induced mice. Figure
9I shows colon tumour incidence, colon tumour number and colon tumour load (total
tumour area, mm2) (Figure 9J) of AOM-induced mice. Figure 9K shows representative H & E staining images
(scale bar=200µm) and Ki-67 IHC staining (scale bar=50µm) of AOM-induced mice.
Figures 10A - 10B illustrate safety assessment of R. intestinalis on APCMin/+ mice. The dosage and frequency of R. intestinalis treatment on mouse body weight (Figure 10A), live (ALT, AST) and renal function (BUN) (Figure 10B). ALT, Alanine transaminase; AST, aspartate aminotransferase; BUN, blood urea nitrogen;
CREA, creatinine, all the samples were under 0.3mg/dL.
Figures 11A - 11C show R. intestinalis enhances epithelial barrier functions in ApcMin/+ mice. Figure 11A shows FITC-dextran (4KD) concentration (ng/ml) in ApcMin/+ mice serum. Figure 11B shows expression levels of tight junction proteins ZO-1 and
Claudin-3 determined by western-blot in colon tissues of ApcMin/+ mice. Figure 11C is representative ZO-1 and Claudin-3 IHC staining images (scale
bar=50µm).
Figures 12A and 12B show alive and pasteurized R. intestinalis have no tumour-suppressive effects in vitro. Both alive R.i (CFU=106/ml) (Figure 12A), and pasteurized (70 °C, 30min) R.i (Figure 12B) had no effects on colonic cell viability.
Figures 13A - 13H illustrates R.i CM exhibits tumour-suppressive effects in vitro. As shown in Figure 13A R.i CM (5%) reduced the colonic cell viability, except NCM460. Escherichia coli conditioned medium (E.i CM) and YCFA were used as control. As shown in Figure 13B R.i CM (5%) suppressed the colony formation of CRC cells. As shown in Figure 13C R.i CM reduced CRC cell Ki-67 expression levels determined by immunocytochemistry (ICC).
As shown in Figure 13D R.i CM increased CRC cell apoptosis. Figure 13E shows R.i CM induced CRC cell apoptosis of CRC cells by cleaved caspase-3, cleaved caspase-7,
cleaved caspase-9, and cleaved PARP expression. Figure 13F shows R.i CM inhibited CRC cell cycle G1/S transition. Figure 13G shows R.i CM induced G1 arrest by Cdk6 downregulation and p27 Kip1 upregulation. Figure 13H
shows R.i CM inhibited the growth of the organoid derived from a CRC patient.
Figures 14A - 14D show the tumour suppressive component produced by R. intestinalis is Butyric acid. As shown in 14A, the efficient components in R.i CM are less than 3KD. As shown in Figure 14B, the R.i CM <3KD fraction was assessed by untargeted LC-MS/MS analysis and butyric acid was
identified as the candidate tumour suppressive molecule. Figure 14C shows butyrate
acid concentration (µg/ml) in R.i CM <3KD was determined by targeted short chain fatty acid GC-MS analysis. Figure
14D shows relative concentration of butyrate acid (area under curve, AUC) in ApcMin/+ and AOM-induced mice feces was determined by GC-MS analysis.
Figures 15A - 15I show comparable dosage of butyrate in R.i CM suppresses colorectal cancer cells by directly antitumour effects and CD8+ T cell boosting in vitro. Figure 15A shows comparable dosage (1mM) of butyrate in R.i CM reduced the colonic cell viability, except NCM460. Figure 15B shows butyrate induced
CRC cell apoptosis. Figure 15C shows butyrate inhibited CRC cell cycle G1/S transition.
Figure 15D shows R.i CM directly boosted Granzyme B+, IFN-γ+, TNF-α+ CD8+ T cells isolated from human PBMC. The mRNA expression level of associated receptors
on CD8+ T cells after treatment with R. i CM is shown in Figure 15E, and Butyrate (Figure 15F). Figure 15G shows the binding
affinity between TLR5 and butyric acid was measured by surface plasmon resonance (SPR)
(left), affinity of TLR5 to butyric acid was 264 µM (right). Figure 15H shows Granzyme
B+, IFN-γ+, TNF-α+ CD8+ T cells isolated from human PBMC were directly boosted by comparable dosage of butyrate
and abolished by TLR5 antagonist TH1020. Figure 15I shows Phospho-NF-κB p65 protein
expression level of αCD3/αCD28/IL2 activated Jurkat E6.1 T cells after R.i CM and butyrate treatment with or without TH1020 inhibition.
Figures 16A and 16B shows Inosine might be another tumour suppressive component produced by R.i. Figure 16A shows the R.i CM <3KD fraction was assessed by untargeted GC-MS/MS analysis and inosine was identified
as the candidate tumour suppressive molecule. As shown in Figure 16B, inosine concentration
(µg/ml) in R.i CM <3KD was determined by targeted LC-MS analysis.
Figures 17A - 17D show the comparable dosage of inosine in R.i CM has no anti-tumour effects in vitro. Comparable dosage of inosine in R.i CM (0.4µg/ml) had no suppressive effects on colonic cell viability (Figure 17A),
cell apoptosis (Figure 17B) and cell cycle (Figure 17C). Comparable dosage of inosine
had no effects on Granzyme B+, IFN-γ+, TNF-α+ CD8+ T cells isolated from C57BL/6 mouse spleen.
Figures 18A and 18B show comparable dosage of butyrate in R.i CM suppresses colorectal cancer cells by CD8+ T cell boosting in vitro. R.i CM (Figure 18A) and comparable dosage (1mM) of butyrate (Figure 18B) directly boosted
Granzyme B+, IFN-γ+, TNF-α+ CD8+ T cells isolated from C5TBL/6 mouse spleen.
Figures 19A - 19G show R. intestinalis and butyrate restrict MC38 orthotopic tumour growth by directly boosting CD8+ T cells. Figure 19A is a schematic diagram showing the experimental design and timeline of
MC38 orthotopic model. Figure 19B shows representative MC38 rectum orthotopic tumour
morphologies. Figure 19C shows tumor volume (mm3) and Figure 19D shows tumour weight. Figure 19E shows the proportion of tumour infiltrating
CD8+ T cells in all nucleated cells of MC-38 orthotopic tumour tissues determined by IHC.
Figure 19F shows frequencies of granzyme B, IFN-γ, TNF-α within CD8+ populations of
tumours from control, E. coli and R.i treated C57BL/6J mice. Figure 19G shows the frequencies of MDSC from tumour tissues
in control, E. coli and R.i treated C57BL/6J mice measured by flow cytometry.
Figures 20A - 20G show R. intestinalis and butyrate treatments are effective in CT-26 orthotopic tumour via CD8+ T cells independently from anti-PD-1 therapy. Figure 20A is a schematic diagram showing the experimental design and timeline of
CT26 orthotopic model. Figure 20B shows representative CT26 rectum orthotopic tumour
morphologies. Figure 20C shows tumour volume (mm3) and Figure 20D shows tumour weight. Figure 20E shows a proportion of tumour infiltrating
CD8+ T cells of CT-26 orthotopic tumour in all nucleated cells determined by IHC. Figure
20F shows frequencies of granzyme B, IFN-γ, TNF-α within CD8+ populations of tumours
from control, E. coli and R.i treated BALB/c mice. Figure 20G shows frequencies of MDSC from tumour tissues in
control, E. coli and R.i treated BALB/c mice measured by flow cytometry.
Figures 21A - 21C show only S. salivarius K12 inhibits the viability of colon cancer cells. Figure 21A shows the viability of CRC cells (HCT116, Lovo) and normal colon cells
(NCM460) after co-incubation with or without 3 different strains of S. salivarius (K12, M18, Human). E. coli was used as a bacteria control. Figure 21B shows the viability of CRC cells (HCT116,
Lovo) and normal colon cells (NCM460) after co-incubation with or without the conditioned
medium of 3 different strains of S. salivarius (K12, M18, Human). The conditioned medium of E. coli (Ec.CM) was used as a bacteria control. BHI was used as broth control. Figure 21C
shows the genome tree form of 3 different strains of S. salivarius (K12, M18, Human), and the specific EPS gene cluster from S. salivarius K12. Results are presented as mean ± S.D. Statistical significance was determined
by one-way ANOVA or two-way ANOVA where appropriate. S.s, Streptococcus salivarius; CM, conditioned medium; MOI, multiplicity of infection; EPS, exopolysaccharides.
Figures 22A - 22D show S. salivarius K12 protects against intestinal tumourigenesis and modulates anti-tumour immunity
in Apcmin/+ mice. Figure 22A is a schematic diagram showing the experimental design, timeline, and
representative colonic morphologies of Apcmin/+ mice under different treatments. Figure 22B shows colonic, small intestine, and total
(Colon + small intestine) tumour number of Apcmin/+ mice under different treatments. Figure 22C shows colonic, small intestine, and total
(Colon + small intestine) tumour load of Apcmin/+ mice under different treatments. Figure 22D shows the proportion of tumour infiltrating
CD8+ T cells in Apcmin/+ mice under different treatments. Results are presented as mean ± S.D. Statistical
significance was determined by one-way ANOVA. S.s, Streptococcus salivarius; SI, small intestine.
Figures 23A - 23D shows S. salivarius K12 protects against intestinal tumourigenesis and modulates anti-tumour immunity
in AOM/DSS mouse model. Figure 23A is a schematic diagram showing the experimental design, timeline, and
representative colonic morphologies of AOM/DSS-induced CRC mice under different treatments.
Figure 23B shows representative images of colon tumour from AOM/DSS mouse model. Figure
23C shows colon tumour number and tumour size in AOM/DSS mice under different treatments.
Figure 23D shows the proportion of tumour infiltrating CD8+ T cells in AOM/DSS mouse
model under different treatments. Results are presented as mean ± S.D. Statistical
significance was determined by one-way ANOVA. S.s K12, Streptococcus salivarius K12; AOM, azoxymethane.
Figures 24A - 24D shows S. salivarius K12 protects against intestinal tumourigenesis and modulates anti-tumour immunity
in the MC38 syngeneic mouse model. Figure 24A is a schematic diagram showing the experimental design, and timeline of
MC38 syngeneic mouse model under different treatments. Figure 24B shows representative
images of tumours from MC38 syngeneic mouse model under different treatments. Figure
24C shows tumour size and tumour weight in MC38 syngeneic mouse model under different
treatments. Figure 24D shows the proportion of tumour infiltrating CD8+ T cells in
MC38 syngeneic mouse model under different treatments. Results are presented as mean
± S.D. Statistical significance was determined by one-way ANOVA or two-way ANOVA where
appropriate. S.s K12, Streptococcus salivarius K12.
Figures 25A - 25D shows S. salivarius K12 protects against intestinal tumourigenesis and modulates anti-tumour immunity
in the CT26 syngeneic mouse model. Figure 25A is a schematic diagram showing the experimental design, and timeline of
CT26 syngeneic mouse model under different treatments. Figure 25B shows representative
images of tumours from Ct26 syngeneic mouse model under different treatments. Figure
25C tumour size and tumour weight in CT26 syngeneic mouse model under different treatments.
Figure 25D shows the proportion of tumour infiltrating CD8+ T cells in CT26 syngeneic
mouse model under different treatments. Results are presented as mean ± S.D. Statistical
significance was determined by one-way ANOVA or two-way ANOVA where appropriate. K12,
Streptococcus salivarius K12; aPD1, anti-PD1.
DEFINITIONS
[0009] In this disclosure, the terms "colorectal cancer (CRC)" and "colon cancer" have the
same meaning and refer to a cancer of the large intestine (colon), the lower part
of human digestive system, although rectal cancer often more specifically refers to
a cancer of the last several inches of the colon, the rectum. A "colorectal cancer
cell" is a colon epithelial cell possessing characteristics of colon cancer and encompasses
a precancerous cell, which is in the early stages of conversion to a cancer cell or
which is predisposed for conversion to a cancer cell. Such cells may exhibit one or
more phenotypic traits characteristic of the cancerous cells.
[0010] The term "inhibiting" or "inhibition," as used herein, refers to any detectable negative
effect on a target biological process, such as RNA/protein expression of a target
gene, the biological activity of a target protein, cellular signal transduction, cell
proliferation, presence/level of an organism especially a micro-organism, any measurable
biomarker, bio-parameter, or symptom in a subject, and the like. Typically, an inhibition
is reflected in a decrease of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%
or greater in the target process (e.g., a subject's bodyweight, or the blood glucose/
cholesterol level, or any measurable symptom or biomarker in a subject, such as an
infection rate among subjects by a pathogenic infectious agent), or any one of the
downstream parameters mentioned above, when compared to a control. "Inhibition" further
includes a 100% reduction, i.e., a complete elimination, prevention, or abolition
of a target biological process or signal. The other relative terms such as "suppressing,"
"suppression," "reducing," and "reduction" are used in a similar fashion in this disclosure
to refer to decreases to different levels (e.g., at least 10%, 20%, 30%, 40%, 50%,
60%, 70%, 80%, 90% or greater decrease compared to a control level) up to complete
elimination of a target biological process or signal. On the other hand, terms such
as "activate," "activating," "activation," "increase," "increasing," "promote," "promoting,"
"enhance," "enhancing," or "enhancement" are used in this disclosure to encompass
positive changes at different levels (e.g., at least about 5%, 10%, 20%, 30%, 40%,
50%, 60%, 70%, 80%, 90%, 100%, 200%, or greater such as 3, 5, 8, 10, 20-fold increase
compared to a control level in a target process, signal, or parameter.
[0011] As used herein, the term "treatment" or "treating" includes both therapeutic and
preventative measures taken to address the presence of a disease or condition or the
risk of developing such disease or condition at a later time. It encompasses therapeutic
or preventive measures for alleviating ongoing symptoms, inhibiting or slowing disease
progression, delaying of onset of symptoms, or eliminating or reducing side-effects
caused by such disease or condition. A preventive measure in this context and its
variations do not require 100% elimination of the occurrence of an event; rather,
they refer to a suppression or reduction in the likelihood or severity of such occurrence
or a delay in such occurrence.
[0012] The term "severity" of a disease refers to the level and extent to which a disease
progresses to cause detrimental effects on the well-being and health of a patient
suffering from the disease, such as short-term and long-term physical, mental, and
psychological disability, up to and including death of the patient. Severity of a
disease can be reflected in the nature and quantity of the necessary therapeutic and
maintenance measures, the time duration required for patient recovery, the extent
of possible recovery, the percentage of patient full recovery, the percentage of patients
in need of long-term care, and mortality rate.
[0013] A
"patient" or
"subject" receiving the composition or treatment method of this invention is a human, including
both adult and juvenile human, of any age, gender, and ethnic background, who have
been diagnosed with colon cancer and receiving immunotherapy for the condition and
are therefore in need of enhancing efficacy of the treatment. Typically, the patient
or subject receiving treatment according to the method of this invention to improve
immunotherapy efficacy is not otherwise in need of treatment by the same therapeutic
agents. For example, if a subject is receiving the synbiotic composition according
to the claimed method, the subject is not suffering from any other disease that is
known to be treated by the same therapeutic agents. Although a patient may be of any
age, in some cases the patient is at least 40, 45, 50, 55, 60, 65, 70, 75, 80, or
85 years of age; in some cases, a patient may be between 40 and 45 years old, or between
50 and 65 years of age, or between 65 and 85 years of age. A "child" subject is one
under the age of 18 years, e.g., about 5-17, 9 or 10-17, or 12-17 years old, including
an "infant," who is younger than about 12 months old, e.g., younger than about 10,
8, 6, 4, or 2 months old, whereas an "adult" subject is one who is 18 years or older.
[0014] The term
"immunotherapy," as used in the context of colon cancer treatment, refers to any treatment methodology
that achieves inhibition of cancer cell proliferation, as indicated by tumour mass
increase and/or metastasis, by way of inducing, stimulating, or enhancing a patient's
immune system functions to suppress or kill cancer cells. For example,
anti-PD1 immunotherapy is a frequently used cancer treatment strategy that targets the PD1 and PDL1 interaction
between cancer cells and immune cells. PDL1 (Programmed Cell Death Ligand 1) is a
protein found on the surface of some cancer cells that interacts with the PD1 (Programmed
Cell Death 1) protein on T cells, which are a part of the immune system normally capable
of immune surveillance and initial elimination of newly arisen cancer cells. The PDL1
to PD1 interaction acts to thwart an attack on the cancer cells by a normal immune
system. It's one of the molecular mechanisms that cancer cells are known to rely on
to escape detection and destruction by the immune system and thus to continue their
uncontrolled growth and proliferation. The use of PDL1 or PD1 inhibitors, the so-called
immune checkpoint inhibitors (ICIs), for cancer treatment is therefore known as anti-PD1
immunotherapy for the inhibitors' ability to restore the anti-cancer capability of
a patient's immune system. Although many known PDL1 or PD1 inhibitors are antibodies
(especially human or humanized antibodies) against the PDL1 or PD1 protein, any other
molecules, including small molecules, can also be employed in a PD1 immunotherapy
if they are capable of interacting with either PDL1 or PD1 and therefore disrupting
the interaction between the two proteins.
[0015] The term
"effective amount," as used herein, refers to an amount that produces intended (e.g., therapeutic or
prophylactic) effects for which a substance is administered. The effects include the
prevention, correction, or inhibition of progression of the symptoms of a particular
disease/condition and related complications to any detectable extent,
e.g., incidence of disease, progression rate, severity of disease, including tumour mass,
distant metastasis, and up to mortality. The exact amount will depend on the purpose
of the treatment, and will be ascertainable by one skilled in the art using known
techniques (
see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992);
Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); and
Pickar, Dosage Calculations (1999)).
[0016] The term
"about" when used in reference to a given value denotes a range encompassing ±10% of the
value.
[0017] A
"pharmaceutically acceptable" or
"pharmacologically acceptable" excipient is a substance that is not biologically harmful or otherwise undesirable,
i.e., the excipient may be administered to an individual along with a bioactive agent
without causing any undesirable biological effects. Neither would the excipient interact
in a deleterious manner with any of the components of the composition in which it
is contained.
[0018] The term
"excipient" refers to any essentially accessory substance that may be present in the finished
dosage form of the composition of this invention. For example, the term "excipient"
includes vehicles, binders, disintegrants, fillers (diluents), lubricants, glidants
(flow enhancers), compression aids, colours, sweeteners, preservatives, suspending/dispersing
agents, film formers/coatings, flavours, and printing inks.
[0019] The term
"consisting essentially of," when used in the context of describing a composition containing an active ingredient
or multiple active ingredients, refer to the fact that the composition does not contain
other ingredients possessing any similar or relevant biological activity of the active
ingredient(s) or capable of enhancing or suppressing the activity, whereas one or
more inactive ingredients such as physiological or pharmaceutically acceptable excipients
may be present in the composition. For example, a composition consisting essentially
of active agents (for instance, one or more of
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12) effective for enhancing or improving efficacy of immunotherapy for treating
colon cancer in a subject is a composition that does not contain any other agents
that may have any detectable positive or negative effect on the same target process
(
e.g., anti-colon cancer efficacy) or that may increase or decrease to any measurable
extent of the disease severity or outcome among the receiving subjects.
DETAILED DESCRIPTION OF THE INVENTION
I. Introduction
[0020] This invention describes the use of certain beneficial gut bacterial species (
e.g., one or more of
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12), optionally in further combination with certain prebiotics (
e.g., ICA and butyrate), for enhancing or improving the therapeutic efficacy of an anti-colon
cancer treatment that is concurrently administered to a patient suffering from the
disease, especially for enhancing or improving the efficacy of an immunotherapy such
as the anti-PD1 therapy. The practical use of the invention includes development and
manufacturing of commercial food products or health supplements containing the probiotics
and/or prebiotics, for example, in the form of a powder, tablet, capsule, or liquid,
which can be taken alone or added to food or beverages in connection with anti-colon
cancer treatment efforts at or around the same time.
II. Pharmaceutical Compositions and Administration
[0021] The present invention provides pharmaceutical compositions comprising an effective
amount of one or more of
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12, optionally in further combination with ICA and/or butyrate for enhancing or
improving therapeutic efficacy in a colon cancer patient currently receiving immunotherapy
such as anti-PD1 therapy for treating the disease. Pharmaceutical compositions of
the invention are suitable for use in a variety of drug delivery systems.
[0022] The pharmaceutical compositions of the present invention can be administered by various
routes,
e.g., systemic administration via oral ingestion or local delivery using a rectal suppository.
The preferred route of administering the pharmaceutical compositions is oral administration
at daily doses of about 10
6 to about 10
14 CFU,
e.g., about 5 × 10
9 CFU, for each of one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, at a roughly equal weight ratio, or up to about 3 to 1 weight ratio between
the bacterial species at the highest concentration and the lowest concentration. Optionally,
the prebiotic combination of ICA and butyrate is further administered to the subject,
either in one single composition or in multiple compositions. Typically, ICA and butyrate
may be present in the composition in the weight ratio of about 1 to about 4, about
1 to about 2, about 1 to about 1, about 1 to about 2, or about 1 to about 4. For example,
ICA and/or butyrate may be present in total amount of about 0.1 to about 4 grams,
preferably about 0.6 to about 2.6 grams. Additionally, the composition may be formulated
in a daily dosage format comprising ICA and/or butyrate in the total amount of about
0.01 to about 50 mg/kg, about 0.1 to about 25 mg/kg, about 1 to about 20 mg/kg, or
about 2 to about 10 mg/kg of the patient. The appropriate dose may be administered
in a single daily dose or as divided doses presented at appropriate intervals, for
example as two, three, four, or more subdoses per day. The duration of administration
may range from about 1 or 2 weeks to about 4 or 8 weeks,
e.g., about 1 week to about 2 weeks, or about 1 month to about 2 months, just prior to,
during, or shortly after (for example, within about 1, 2, 3, 4, 5 days or a week before
or after) an immunotherapy (
e.g., anti-PD1 therapy) is administered to the patient.
[0023] For preparing pharmaceutical compositions containing one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally with prebiotics ICA and/or butyrate, one or more inert and pharmaceutically
acceptable carriers may be used. The pharmaceutical carrier can be either solid or
liquid. Solid form preparations include, for example, powders, tablets, dispersible
granules, capsules, cachets, and suppositories. A solid carrier can be one or more
substances that can also act as diluents, flavoring agents, solubilizers, lubricants,
suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating
material.
[0024] In powders, the carrier is generally a finely divided solid that is in a mixture
with the finely divided active component,
e.g., one or more of
L. gallinarum, R. intestinalis, and
S.
salivarius K12, optionally further in combination with ICA and/or butyrate. In tablets, the
active ingredient is mixed with the carrier having the necessary binding properties
in suitable proportions and compacted in the shape and size desired.
[0025] For preparing pharmaceutical compositions in the form of suppositories, a low-melting
wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and
the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous
mixture is then poured into convenient-sized molds and allowed to cool and solidify.
[0026] Powders and tablets preferably contain between about 5% to about 100% by weight of
the active ingredient(s) (
e.g., one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally further in combination with ICA and/or butyrate). Suitable carriers
include, for example, magnesium carbonate, magnesium stearate, talc, lactose, sugar,
pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose,
a low-melting wax, cocoa butter, and the like.
[0027] The pharmaceutical compositions can include the formulation of the active ingredient(s)
e.g., one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally further in combination with ICA and/or butyrate, with encapsulating
material as a carrier providing a capsule in which the active ingredient(s) (with
or without other carriers) is surrounded by the carrier, such that the carrier is
thus in association with the active ingredient(s). In a similar manner, sachets can
also be included. Tablets, powders, sachets, and capsules can be used as solid dosage
forms suitable for oral administration.
[0028] Liquid pharmaceutical compositions include, for example, solutions suitable for oral
administration or local delivery, suspensions, and emulsions suitable for oral administration.
Sterile water solutions of the active component (
e.g., one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally further in combination with ICA and/or butyrate) or sterile solutions
of the active component in solvents comprising water, buffered water, saline, PBS,
ethanol, or propylene glycol are examples of liquid or semi-liquid compositions suitable
for oral administration or local delivery such as by rectal suppository. The compositions
may contain pharmaceutically acceptable auxiliary substances as required to approximate
physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting
agents, wetting agents, detergents, and the like.
[0029] Sterile solutions can be prepared by dissolving the active component (
e.g., one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally further with ICA and/or butyrate) in a previously sterilized solvent
under sterile conditions. In some cases, the composition will consist essentially
of one or more of
L. gallinarum, R. intestinalis, and
S.
salivarius K12, optionally with ICA and/or butyrate, plus one or more physiological excipients,
without another microorganism present at a detectable level, which is defined as undetectable
by means of a polymerase chain reaction. The resulting aqueous solutions may be packaged
for use as is, or lyophilized, the lyophilized preparation being combined with a sterile
aqueous carrier prior to administration. The pH of the preparations typically will
be between 3 and 11, more preferably from 5 to 9, and most preferably from 7 to 8.
[0030] Single or multiple administrations of the compositions can be carried out with dose
levels and pattern being selected by the treating physician. In any event, the pharmaceutical
formulations should provide a quantity of an active agent sufficient to effectively
enhance the efficacy of a vaccine and/or reduce or eliminate undesirable adverse effects
of a vaccine.
III. Additional Therapeutic Agents
[0031] Additional known therapeutic agent or agents may be used in combination with an active
agent such as one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally further in combination with prebiotics such as ICA and/or butyrate,
in the practice of the present invention for the purpose of enhancing or improving
the efficacy of an immunotherapy for treating colon cancer in a patient in need thereof.
In such applications, one or more of these previously known effective anti-cancer
therapeutic agents can be administered to patients concurrently with an effective
amount of the active agent(s) either together in a single composition or separately
in two or more different compositions.
[0032] For example, therapeutic agents that are known to be effective for use to treat cancer,
especially colon cancer, include Camptosar (irinotecan hydrochloride), capecitabine,
Eloxatin (oxaliplatin), 5-fluorouracil, leucovorin calcium, Lonsurf (trifluridine
and tipiracil hydrochloride), Stivarga (regorafenib), Tukysa (tucatinib), Zaltrap
(ziv-aflibercept), and their combinations may be used along with the active agents
(such as the probiotics, optionally with the prebiotics) of the present invention
to enhance an immunotherapy's effectiveness, reduce potential disease severity (including
morbidity and mortality), prevent/reduce risk of metastasis or recurrence, and improve
patient survival and recovery from the disease.
IV. Kits
[0033] The invention also provides kits for improving therapeutic efficacy in an individual
receiving immunotherapy for the treatment of colon cancer according to the method
disclosed herein. The kits typically include a plurality of containers, each containing
a composition comprising one or more of beneficial bacterial species
Lactobacillus gallinarum, Roseburia intestinalis, and
Streptococcus salivarius K12. Optionally, additional container(s) may be included in the kit providing composition(s)
comprising ICA or butyrate or both. Further, additional agents or drugs that are known
to be therapeutically effective for treating colon cancer, including for ameliorating
the symptoms and reducing the severity of the disease, as well as for facilitating
survival/recovery from the disease (such as those described in the last section or
otherwise known in the pertinent technical field) may be included in the kit. The
plurality of containers of the kit each may contain a different active agent/drug
or a distinct combination of two or more of the active agents or drugs. Also included
in the kit, in some cases, is one or more compositions containing agent(s) used in
immunotherapy for treating colon cancer, such as any one of PD1 inhibitors (
e.g., Pembrolizumab, Nivolumab, Cemiplimab, Dostarlimab, Vopratelimab, Spartalizumab, Camrelizumab,
Sintilimab (IBI308), Tislelizumab, Toripalimab and Acrixolimab) or PDL1 inhibitors
(
e.g., Atezolizumab, Avelumab, Durvalumab, and Cosibelimab) or any combination thereof.
[0034] The kit may further include informational material providing instructions on how
to dispense the pharmaceutical composition(s), including description of the type of
patients who may be treated (
e.g., human patients who have been diagnosed with colon cancer and are therefore receiving
immunotherapy to treat the disease, such as anti-PD 1 therapy), and in some cases
information about the type of patients not to be included in the claimed method (
e.g., those who have been diagnosed with a pre-existing condition that already requires
the administration of the active components such as one or more of
L. gallinarum, R. intestinalis, and
S. salivarius K12, optionally along with ICA and/or butyrate), as well as the dosage, frequency,
and manner of administration, and the like.
EXAMPLES
[0035] The following examples are provided by way of illustration only and not by way of
limitation. Those of skill in the art will readily recognize a variety of non-critical
parameters that could be changed or modified to yield essentially the same or similar
results.
EXAMPLE 1: Lactobacillus gallinarum and Its Derived Metabolite Improve Anti-PD1 Efficacy in Colorectal Cancer by Modulating
the IDO1/Kyn/AHR Metabolic Zxis
BACKGROUND OF THE INVENTION
[0036] The introduction of immune checkpoint blockade (ICB) therapy has revolutionized the
paradigm of cancer treatment over the past decade. Harnessing the immune system, ICB
therapy removes the "brake" signal of T cell activation and elicits antitumour response.
ICB therapy, namely anti-PD1/PDL1 (programmed cell death protein 1/ programmed death
ligand 1) and anti-CTLA4 (cytotoxic T-lymphocyte-associated protein 4), has yielded
great success in clinical trials and is now approved to treat different cancers. Nevertheless,
in colorectal cancer (CRC), clinical use of ICB therapy remains highly limited and
is only approved in CRC patients with high level microsatellite instability (MSI-H)
or mismatch repair deficiency, making up merely 15% of CRC patients. Meanwhile, drug
resistance persists as an unresolved challenge - a large portion of patients, despite
their eligibility to receive ICB, may be refractory or even fail to respond to anti-PD1
therapy. Therefore, there is a dire need to identify novel adjuvants to overcome ICB
resistance and improve treatment efficacy.
[0037] Gut microbiota has emerged as an important factor dictating ICB response in various
cancers. By using metagenomic profiling, several studies reported a distinct microbiota
composition between ICB responders and non-responders in melanoma patients, while
supplementing bacteria species enriched in responders or faecal microbiota transplantation
to non-responders dramatically improved their ICB efficacy in patients with in melanoma
or epithelial cancer. A recent study also demonstrated that a consortium of 11 bacterial
strains from healthy donors effectively induced IFNγ+ CD8+ T cells, which boosted
antitumour immunity and synergized with ICB therapy.
[0038] While earlier studies focus on the correlation between microbiota and host response,
the immunoregulatory molecules/components and mechanisms remain largely unknown. In
view of this, recent publications have characterized the bacteria-derived immunomodulatory
components, which are mostly bacteria metabolites or cell wall fragments such as inosine,
exopolysaccharides, phospholipids and peptidoglycan. These have highlighted the potential
of leveraging probiotics or bacteria-based therapeutics as a next-generation adjuvant
to improve cancer immunotherapy.
[0039] We demonstrated that
Lactobacillus gallinarum, a probiotic depleted in CRC patients, inhibited colorectal tumourigenesis by secreting
indole-3-lactic acid (ILA), a tryptophan metabolite. Tryptophan metabolites are known
as a class of immunoregulatory metabolites, which could induce Th17 polarization and
promote differentiation of CD4+CD8αα+ double-positive intraepithelial lymphocytes.
Therefore, we sought to explore the translational potential of this probiotic beyond
cancer prevention - whether
L. gallinarum may modulate antitumour immunity and improve ICB treatment in CRC. Here, we report
that
L. gallinarum-derived indole-3-carboxylic acid (ICA) significantly improved anti-PD1 therapy in
CRC. We revealed that ICA reduced regulatory T cell (Treg) infiltration and enhanced
the effector function of CD8+ T cells, mechanistically by modulating the indoleamine
2,3-dioxygenase 1 (IDO1)/ Kynurenine (Kyn)/ aryl hydrocarbon receptor (AHR) metabolic
axis.
MATERIALS AND METHODS
Bacteria strains and growth conditions
[0040] L. gallinarum (ATCC 33199) and
E. coli strain MG1655 (ATCC 700926) were purchased from American Type Culture Collection
(ATCC; Manassas, VA).
E. coli strain MG1655 is a non-pathogenic human commensal gut bacterium and was used as a
negative control in this study.
L. gallinarum and
E. coli were cultured in MRS or BHI broth as appropriate in a shaking incubator at 37°C under
aerobic condition, and were resuspended in BHI broth before gavage.
Cell culture
[0041] Murine MC38 (microsatellite instability high, MSI-H) and CT26 (microsatellite stable,
MSS) and human (HCT116 and LoVo) CRC cell lines were purchased from ATCC. All cells
were cultured in Dulbecco's modified Eagle's medium (Gibco BRL, Grand Island, NY)
supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin, and
maintained at 37°C in a humidified incubator with 5% carbon dioxide.
Syngeneic mouse model
[0042] Male C57BL/6 or BALB/c mice at 5-6 weeks old were randomized and daily gavaged with
(1) brain heart infusion (BHI) broth, (2)
Escherichia coli MG1655, or (3)
Lactobacillus gallinarum (1×10
8 colony-forming unit/100µL per mouse). After one week, MC38 (5×10
5 cells) or CT26 (1×10
6 cells) were subcutaneously injected into the dorsal flank of C57BL/6 and BALB/c mice,
respectively. When the tumour size reached 50-100 mm
3, anti-mouse programmed cell death protein 1 (anti-PD1) monoclonal antibody (BE0146,
Bio X Cell, Lebanon, NH) or IgG isotype control (BE0089, Bio X Cell) was administered
to mice by intraperitoneal injection every 3 days (100µg per mouse). Tumour size was
measured by caliper every other day.
[0043] For indolecarboxylic acid (ICA) model, ICA (18mg/kg, Sigma-Aldrich, St. Louis, MO)
was gavaged to mice daily. Tumour inoculation and dosing schedule of anti-PD1 or isotype
control is the same as described above. After the outgrowth of palpable tumours, kynurenine
(Kyn, 10mg/kg, Sigma-Aldrich) was intraperitoneally injected into mice twice weekly.
Carcinogen-induced colon cancer model
[0044] Male C57BL/6 mice at 5-6 weeks old were intraperitoneally injected with a single
dose of azoxymethane (AOM; Merck, Darmstadt, Germany), followed by 1 week of 1% dextran
sulphate sodium (DSS; MP Biomedicals, Solon, OH) in drinking water and a 2-week recovery
period. A total of 3 DSS cycles were given. After the third DSS cycle, the mice were
randomized and gavaged with BHI broth,
E. coli or
L. gallinarum daily. Anti-PD1 or isotype control (100µg) were given to mice by intraperitoneal
injection every 3 days after one week of bacterial gavage. Mouse colonoscopy (Karl
Storz Endoskope, Tuttlingen, Germany) was performed before sacrifice. All animal studies
were approved by the Animal Experimentation Ethics Committee of The Chinese University
of Hong Kong.
Metabolomics profiling and metabolites analysis
[0045] Metabolomics was performed by BIOTREE, Shanghai, China. The metabolites were transferred
to a fresh glass vial for liquid chromatography-tandem mass spectrometry (LC-MS/MS)
analysis. Ultra-high performance liquid chromatography (UHPLC) separation was carried
out using a 1290 Infinity series UHPLC System (Agilent Technologies, Palo Alto, CA),
equipped with a UPLC BEH Amide column. The TripleTOF 6600 mass spectrometry (AB Sciex,
Foster City, California) was used to acquire tandem mass spectrometry (MS/MS) spectra
on an information-dependent basis during LC-MS/MS experiment. The metabolite identification
was based on in-house MS2 database, Human Metabolome Database (HMDB, website: hmdb.ca),
and METLIN metabolite database (metlin.scripps.edu).
Multicolor flow cytometry analysis
[0046] Tumour tissues were dissected into small pieces and digested with 1% bovine serum
albumin (BSA) containing 0.5mg/ml collagenase D (Roche, Basel, Switzerland) and 0.25mg/ml
DNase I (Roche) for 45 minutes at 37°C on a shaking platform. The cell suspension
was then filtered through a 70-µm cell strainer and centrifuged at 700 g for 10 minutes.
The cell pellets were resuspended in 1% BSA for surface marker staining. Nuclear antigen
staining (Foxp3) was performed using Foxp3 staining buffer set (eBioscience, San Diego,
CA) according to manufacturer's instructions. For intracellular cytokine staining,
the cell suspension was first stimulated with phorbol myristate acetate, ionomycin
and monensin for 4 hours at 37°C before surface marker staining. Then, the cells were
fixed and permeabilized by Foxp3 staining buffer (eBioscience), followed by antibody
staining.
Immunohistochemistry staining
[0047] Formalin-fixed, paraffin-embedded tissues were sectioned in 4µm thickness. Heat-mediated
antigen retrieval was performed using 10mM sodium citrate buffer (pH 6.0) for 20 minutes.
Immunohistochemistry (IHC) staining was conducted using the IHC Select HRP/DAB kit
(Millipore, Burlington, MA) according to manufacturer's instructions. Tissue sections
were stained with IDO1 antibody (ab106134, Abcam, Cambridge, UK) at 1:100 dilution.
T cell isolation and in vitro experiments
[0048] T cells were isolated from spleens of tumour-bearing mice by negative selection using
magnetic cell separation (EasySep mouse T cell isolation kit, CD8+ T cell isolation
kit or CD4+ CD25+ Regulatory T cell Isolation Kit II; STEMCELL Technologies, Vancouver,
Canada). Isolated T cells were cultured in Roswell Park Memorial Institute 1640 medium
supplemented with 10% FBS, 50 µM β-mercaptoethanol, 10ng/mL mouse recombinant interleukin-2
(IL-2), 0.1ng/mL transforming growth factor β (TGFβ) and activated with Dynabeads
mouse anti-CD3/CD28 beads (Thermo Fisher Scientific, Waltham, MA) for 72 hours. Cells
were then collected for analysis.
Reverse transcription-quantitative PCR (RT-qPCR)
[0049] Total RNAs were extracted from cell pellets or colonic tissues using TRIzol Reagent
(Life Technologies). Complementary DNA (cDNA) was prepared using Prime Script RT Reagent
Kit with gDNA Eraser (Takara, Shiga, Japan). The relative level of specific genes
was determined by QuantStudio
™ 7 Flex Real-Time PCR System (Thermo Fisher Scientific). All gene expression values
were normalized to β-actin and calculated using the 2-ΔΔCt method.
Statistical analysis
[0050] Values are expressed as mean ± standard deviation (SD) for both
in vivo and
in vitro experiments. Comparisons between two groups were performed using a two-sided Student's
t-test. Analysis of variance (ANOVA) was used to compare differences among multiple
groups, and post-hoc analysis was performed by Tukey's multiple comparisons test.
P-values < 0.05 indicate statistical significance.
RESULTS
L. gallinarum improved anti-PD1 efficacy in syngeneic mouse models
[0051] To investigate whether
L. gallinarum supplementation improves anti-PD1 efficacy, we first established a syngeneic mouse
model by inoculating a murine MSI-H CRC cell line (MC38) into mice with daily gavage
of either BHI (broth control),
E. coli (bacterial negative control) or
L. gallinarum (Figure 1A). Once tumours were developed, anti-PD1 antibody or IgG isotype control was intraperitoneally
injected to mice.
L. gallinarum remarkably improved anti-PD1 efficacy, as tumour growth was significantly more impeded
by the combination of anti-PD1 and
L. gallinarum, compared to anti-PD1 combined with BHI or
E. coli control (both
P < 0.01)
(Figure 1B -
D).
[0052] MC38 syngeneic model is a highly immunogenic model with good immunotherapy response.
We next sought to examine whether
L.
gallinarum could also be effective in a murine MSS CRC cell line (CT26), a model with low microsatellite
stability (MSI-low) and inherited immunotherapy resistance. As expected, anti-PD1
combined with BHI or
E. coli showed minimal effects on tumour growth in CT26 syngeneic mice
(Figure 1E -
G). In comparison,
L. gallinarum supplementation significantly improved anti-PD1 efficacy, and the combination of
anti-PD1 and
L.
gallinarum induced a marked reduction in tumour weight (
P < 0.01)
(Figure 1E & F) and volume (
P < 0.0001)
(Figure 1G). Altogether, these results indicate that
L.
gallinarum enhanced anti-PD1 efficacy to suppress tumour growth.
L. gallinarum reduced Treg infiltration and enhanced effector function of CD8+ T cells
[0053] Anti-PD1 efficacy is closely associated with immune cell infiltration. We therefore
performed multicolor flow cytometric analysis and examined the change of immune landscape
induced by
L. gallinarum. In MC38 syngeneic mouse model,
L. gallinarum monotherapy, despite its inefficacy in impeding tumour growth, reduced Foxp3+ CD25+
Treg infiltration, compared to BHI and E. coli group (both P < 0.05)
(Figure 2B). In mice receiving
L.
gallinarum plus anti-PD1 therapy, we observed a consistent reduction of Foxp3+ CD25+ Treg infiltration
(both P < 0.05)
(Figure 2B), and also a significant increase of IFNγ+ CD8+ T cells (P < 0.01 for BHI, P < 0.001
for E. coli)
(Figure 2E), which is an indicator of improved CD8+ effector T cell function and strengthened
antitumour immunity. We then validated our findings in the CT26 syngeneic mouse model.
Consistently,
L. gallinarum reduced Treg infiltration, both in monotherapy (both P < 0.05) and combination with
anti-PD1 therapy (both P < 0.01)
(Figure 2A and 2C), and enhanced IFNγ+ CD8+ T cells (both P < 0.001) compared to BHI and E. coli controls
(Figure 2D and 2F).
L. gallinarum sensitized mice to anti-PD1 therapy in CRC tumourigenesis induced by AOM/DSS
[0054] To explore the translational potential of
L. gallinarum in improving immunotherapy, we next established a carcinogen AOM/DSS-induced CRC
tumourigenesis mouse model, which could mimic physiological CRC development in humans.
We initiated
L. gallinarum supplementation after the completion of three DSS cycles, one week before the start
of anti-PD1 treatment, so as to better explore the role of
L. gallinarum as a potential adjuvant to anti-PD1
(Figure 3A). Consistent with syngeneic mouse models, combining
L. gallinarum and anti-PD1 significantly inhibited tumour formation as evidenced by colonoscopy
images prior to sacrifice
(Figure 3B), and the representative colon images after sacrifice
(Figure 3C). Compared to anti-PD1-treated mice receiving BHI or
E. coli control,
L. gallinarum plus anti-PD1 therapy significantly reduced tumour number (both
P < 0.01)
(Figure 3D), tumour load (both
P < 0.05)
(Figure 3E), and number of large tumours (diameter ≥ 2mm) (
P < 0.05 for BHI;
P < 0.01 for
E. coli)
(Figure 3F), indicating
L. gallinarum boosts the efficacy of anti-PD1 therapy in CRC tumourigenesis. Colonic tumours were
collected for immune cell analysis by flow cytometric. Consistent with the findings
from the MC38 and CT 26 allograft models, compared to anti-PD1-treated mice plus BHI
or
E. coli control, the combination of
L. gallinarum and anti-PD1 reduced Treg infiltration (
P < 0.01 for
E. coli;
P < 0.001 for BHI)
(Figure 3G) and enhanced CD8+ effector T cell function (both
P < 0.05)
(Figure 3H). Collectively,
L. gallinarum synergizes with anti-PD1 therapy in CRC treatment in mice.
L. gallinarum produced tryptophan as potential functional metabolite in vitro and in vivo
[0055] We further characterized the metabolomic profile of
L. gallinarum to identify functional metabolites that could modulate antitumour immunity. Untargeted
metabolomic profiling revealed the significant enrichment of multiple tryptophan metabolites,
including indole-3-carboxaldehyde (IAld), indoleacetic acid (IAA) and indolelactic
acid (ILA) in
L. gallinarum culture supernatant
(Figure 4A). In the stool samples of our MC38 syngeneic mouse model, we consistently observed
an elevated level of IAld in
L. gallinarum-treated mice compared to BHI and
E.
coli-treated mice
(Figure 4B). For validation, we then performed targeted metabolomic profiling on 32 different
tryptophan-related metabolites and confirmed the elevated IAld level in stool samples
(Figure 4C). These results incdicate that
L.
gallinarum secreted tryptophan metabolites, specifically lAld, both
in vitro and
in vivo.
[0056] Given that the tumours of syngeneic mice were located at their dorsal flank which
were distant from the intestines, we hypothesized that
L. gallinarum-derived metabolites were first produced in the gut, then entered the bloodstream and finally
reached the tumour tissue. No significant difference in serum lAld level was found
between groups
(Figure 4D), however, indicating that lAld may not be the endpoint metabolite modulating antitumour
immunity. In contrast, a significant enrichment of indole-3-carboxylic acid (ICA),
the downstream metabolite of IAld
20, was observed in serum samples of
L. gallinarum-treated mice, compared to controls (both
P < 0.05)
(Figure 4E). Therefore, we postulated that
L. gallinarum first produced lAld, the precursor of ICA in the gut, which was then converted to
ICA in the bloodstream. ICA may be the functional metabolite that played an immunoregulatory
role.
L. gallinarum and ICA inhibited tumour tissue IDO1 expression
[0057] In our metabolomic panel of tryptophan metabolites, apart from elevated ICA level,
we also observed a decreased Kyn level in both serum (both
P < 0.05) and tumour tissues (both
P < 0.01) of MC38 syngeneic mice with
L.
gallinarum treatment, compared to controls
(Figure 5A and 5B). A decrease of kynurenine-to-tryptophan (Kyn/Trp) ratio in tumour tissues of
L. gallinarum-treated mice was also identified (
P < 0.01 for BHI;
P < 0.05 for
E.
coli)
(Figure 5C). Kyn is a host-derived metabolite notorious for promoting Treg development and immune
evasion, and is primarily produced by indoleamine 2,3-dioxygenase (IDO) through tryptophan
conversion
21. Based on these findings, we hypothesize that
L. gallinarum suppresses IDO expression and/or activity in tumours, which subsequently reduces
Kyn production.
[0058] We therefore performed RNA sequencing on tumour tissues from MC38 syngeneic mouse
model
(Figure 5D). The tryptophan metabolism pathway was significantly downregulated in
L. gallinarum plus anti-PD1 group, compared to BHI plus anti-PD1 and
E.
coli plus anti-PD1 groups (both
P < 0.05)
(Figure 5E). We then performed qPCR to validate the sequencing results, and confirmed in both
MC38 and CT26 syngeneic mouse models that IDO1 expression in tumour tissues was significantly
reduced in
L. gallinarum-treated mice
(Figure 5F). Immunohistochemical (IHC) staining also confirmed the reduced IDO1 expression in
tumour tissues (
P < 0.01 for
E. coli, P < 0.001 for BHI)
(Figure 5G). These results suggest that
L. gallinarum suppressed IDO1 expression in tumour tissues of syngeneic mouse model and reduced
Kyn production.
[0059] Given that ICA was identified as the functional metabolite of
L. gallinarum (Figure 4), we postulated that ICA could be an IDO1 inhibitor. We added IFNγ (100ng/mL) to induce
IDO1 expression. Indeed, ICA significantly suppressed IDO1 expression in two human
CRC cell lines (HCT116 and LoVo)
(Figure 5H). Taken together, these results indicate that
L.
gallinarum or ICA inhibits IDO1 expression and reduces Kyn production in tumour.
ICA antagonized Kyn-mediated Treg differentiation by inhibiting AHR activation
[0060] To explore the impact of
L. gallinarum-derived metabolites on Treg differentiation, we treated naive CD4+ T cells with
L. gallinarum culture supernatant, lAld or ICA. However, none significantly affected Treg differentiation
was observed by each of these treatments, hinting an indirect effect of ICA on Treg
differentiation
(Figure 6A - C). ICA and other tryptophan metabolites are known to activate AHR, while AHR activation
is associated with Treg differentiation and immune escape. Therefore, ICA may compete
with Kyn, a host-derived AHR agonist highly expressed in tumour tissues, and consequently
antagonized Kyn-mediated AHR activation. We simultaneously treated CD4+ T cells with
Kyn and ICA. Kyn (50µM) significantly induced Treg differentiation (
P < 0.0001), while the addition of ICA, at a much lower dose (as low as 1.25µM), antagonized
Kyn-mediated Treg differentiation in a dose-dependent manner
(Figure 6D). We then measured the expression of AHR target gene CYP1B1 on CD4+ T cells. ICA alone
weakly promoted CYP1B1 expression, indicating ICA as a weak AHR agonist. However,
the addition of ICA to Kyn markedly suppressed Kyn-induced CYP1B1 expression
(Figure 6E). The use of CH-233191, an AHR antagonist, also abolished both Kyn and ICA effects
in mediating Treg differentiation
(Figure 6F).
[0061] We also performed the surface plasma resonance (SPR) assay to explore the interaction
between ICA/Kyn with the human AHR receptor. Both ICA and Kyn binds to the human receptor,
whereas ICA has a higher receptor affinity (KD value ~39.4µM) to AHR receptor than
Kyn (61.0 µM)
(Figure 6G). These support our hypothesis that ICA displaces Kyn from AHR receptor and inhibited
Kyn-mediated Treg differentiation through receptor competition.
ICA phenocopied L. gallinarum effect in vivo, reversed by Kyn supplementation
[0062] To confirm the effect of ICA in modulating antitumour immunity
in vivo, we established a syngeneic mouse model with daily gavage of ICA with or without anti-PD1.
ICA significantly promoted anti-PD1 efficacy and phenocopied the effect of
L. gallinarum, as evidenced by the reduced tumour weight and tumour volume, compared to vehicle
control, while Kyn supplementation reversed the effect of ICA
(Figure 7A-C). Consistent with the results of
L. gallinarum-treated mice
(Figure 2), ICA reduced Treg infiltration and increased IFNγ+ CD8+ T cells, which were also reversed
by Kyn supplementation
(Figure 7D&E). ICA also significantly reduced Kyn level in both serum and tumour
(Figure 7F-G), as well as the Kyn/Trp ratio in tumour
(Figure 7H).
[0063] Collectively, our results demonstrate that
L. gallinarum and its derived ICA improve anti-PD1 therapy in a two-fold mechanism: (1) inhibiting
IDO1 expression and Kyn production, (2) inhibiting Kyn binding on AHR receptor through
receptor competition.
DISCUSSION
[0064] In this study, we report that
L. gallinarum supplementation sensitizes mice to anti-PD1 therapy to both syngeneic mouse model
and CRC tumourigenesis mouse model. We identified ICA, a tryptophan metabolite secreted
by
L. gallinarum, as the key player in remodeling TME through modulating the IDO1/Kyn/AHR metabolic
axis. On top of other publications, our results indicate a novel mechanism of how
a probiotic species modulates antitumour immunity and improved ICB response.
[0065] We first demonstrated that
L. gallinarum significantly augmented anti-PD1 efficacy and induced a remarkable tumour shrinkage.
Accumulating evidence suggested that gut microbiota composition shapes the outcome
of cancer immunotherapy, whilst microbiota modulation arises as a promising approach
to improve ICB response. For example, a recent study reported that probiotic
Lactobacillus acidophilus improved ICB efficacy by reducing intratumoural Treg and enhanced effector CD8+ T
cells. Similarly, we revealed that
L.
gallinarum reduced Foxp3+ CD25+ Treg infiltration in TME and enhanced effector function of CD8+
T cells when combined with anti-PD1, indicating the strengthened antitumour immunity.
Notably,
L. gallinarum was effective in improving anti-PD1 efficacy in CT26 syngeneic mouse model, an MSI-L
model known to have minimal response to immunotherapy. Treg recruitment and infiltration
is one of the key mechanisms accounting for ICB resistance, which contributed to the
immunosuppressive TME and prevented the cytotoxic activities of effector T cells.
Meanwhile, to better mimic human CRC development, we also established a carcinogen
AOM/DSS-induced CRC mouse model and revealed that
L. gallinarum also significantly improved anti-PD1 therapy with a marked reduction of tumour number
and load. Taken together, our results from different mouse models suggested an immense
translational potential of leveraging probiotic
L. gallinarum in reshaping the immunosuppressive TME and reversing ICB resistance in clinical practice.
[0066] Through metabolomic profiling, we identified ICA as the functional immunomodulatory
component that improved anti-PD1 therapy. We initially focused on lAld, given its
enrichment in
L. gallinarum culture supernatant and stool samples of
L. gallinarum-treated mice. However, metabolomics revealed no significant difference in serum lAld
level, but a significant enrichment of ICA, the downstream metabolite of IAld, between
L. gallinarum and control groups. Therefore, we reasoned that
L. gallinarum first produced lAld, which was further converted to ICA after entering the bloodstream,
and ICA functioned as the endpoint metabolite that modulated antitumour immunity.
Our previous work demonstrated that
L.
gallinarum produced L-tryptophan, while ILA, its downstream catabolites, exerted anticancer
effect and inhibited colorectal tumourigenesis. Concerning the highly dynamic nature
of microbial tryptophan metabolism, we posited that the tryptophan metabolism pathway
may be diverted to a specific pathway
in vivo, probably subject to multiple factors including species, disease model and treatment
model, therefore accounting for the discrepancy of metabolites identified. Of note,
the tryptophan metabolite family shares many common biological features, such as binding
to AHR, improving gut barrier function and regulating inflammation. In view of this,
we reasonably believe that ICA is not the sole metabolite that modulated antitumour
immunity, but many different tryptophan metabolites may also play a role in remodeling
tumour microenvironment. We also acknowledge that indole metabolites can be produced
by a wide variety of gut bacteria. Therefore, instead of pinpointing
L. gallinarum as the "star" probiotics, it is likely that many gut bacteria also promotes ICB response
through a similar mechanism.
[0067] We identified that
L. gallinarum and its derived ICA regulated the host tryptophan metabolism pathway and inhibited
IDO1 expression in tumour tissues, leading to a reduced production of Kyn, a host-derived
metabolite contributing to tumour progression, immune escape, and Treg development.
The conversion of tryptophan to Kyn is regulated by IDO1, which is highly expressed
in multiple human cancers and has been an attractive pharmaceutical target. In line
with our observations, several studies reported the role of probiotics in inhibiting
IDO1 expression and lowering Kyn concentration
in vivo. Kyn/Trp ratio is also known to be a prognosis marker predicting ICB response. Here
we report an effector mechanism of how gut microbiota modulated ICB response through
host-bacteria metabolic crosstalk. In fact, tryptophan metabolism involves three major
pathways, which are modulated by host, epithelial/ immune cells and enterochromaffin
cells respectively. It is also known that modulation of one of these metabolic pathways
profoundly disturb the others. Collectively, our results indicate that
L. gallinarum and its derived ICA, exert the immunomodulatory effect, at least in part, by modulating
the host tryptophan metabolism pathway and inhibiting Kyn production in the TME.
[0068] Apart from interfering with the upstream IDO1 enzyme, a downstream mechanism was
also reported involving a receptor competition between ICA and Kyn for the AHR binding
site of T cells, which subsequently inhibited Treg differentiation. We first found
that
L.
gallinarum-derived ICA or lAld had insignificant immunomodulatory effect on Treg differentiation,
indicating that ICA improves ICB response through an indirect manner. Indeed, our
results confirm that ICA can suppress Kyn-mediated Treg differentiation through an
AHR-dependent manner. Tryptophan metabolites including both indoles and Kyn are widely
reported as AHR binding ligands. Several studies reported the antagonistic activity
of indole metabolites - while indole metabolites alone function as a weak AHR agonists,
they also exhibited antagonistic activity and inhibited agonist-induced AHR activation.
AHR agonists or indole metabolites are known to play a pleotropic role in cancer.
While AHR agonists alleviated colonic inflammation and thus potentially prevented
colitis-associated CRC, they also promoted immune evasion in TME in the context of
cancer. In a recent publication,
Lactobacillus-derived indole metabolites were reported to drive immunosuppression by activating
AHR on tumour-associated macrophages to promote tumour progression. In our study,
we demonstrated a competitive nature between host- and microbiota-derived AHR ligands,
showing that
L. gallinarum-derived ICA (a weak AHR agonist) outcompeted Kyn and inhibited Kyn-mediated AHR activation.
In fact, the use of partial agonist is not a new concept in drug development. Several
drug classes in clinical use today, such as opioids (for chronic pain relief) and
beta blockers (for treating cardiovascular disease), also include members with partial
agonistic activity. Partial agonists bind and activate receptors, but at a much weaker
extent compared to a full agonist. In this regard, they prevented full receptor activation,
meanwhile maintaining physiological homeostasis. Our results collectively indicate
that gut microbiota-derived AHR ligands function as a partial AHR agonist and suppress
AHR activation in TME in the presence of Kyn, a potent AHR agonist.
[0069] In conclusion, we have demonstrated that
L. gallinarum and its derived ICA can improve anti-PD1 efficacy in CRC. Such actions are associated
with the inhibition of IDO1/Kyn metabolic circuit, as well as the antagonism of Kyn
binding on AHR receptors on T cells to inhibit Treg differentiation. Our results reveal
a novel mechanism underlining host-microbiota metabolic crosstalk and highlight the
translational potential of leveraging
L.
gallinarum as an adjuvant therapy to improve ICB response.
EXAMPLE 2: Butyrate Producing Bacteria Roseburia intestinalis Mediates Cytotoxic cd8+ T Cell Immunity against Colorectal Cancer
BACKGROUND OF THE INVENTION
[0070] Colorectal cancer (CRC) is the third major cancer type and remains the third guiding
cause of cancer-related death worldwide. Despite significant clinical responses to
immunological checkpoint inhibitors in melanoma and non-small cell lung cancer patients,
the role of immunotherapy on CRC treatment is limited. Clinical evidence demonstrated
that only microsatellite instable (MSI) CRC patients responded to PD-1 inhibitor,
while only 10-15% of CRC tumours are MSI. The treatment of most microsatellite stable
(MSS) CRC patients leaves a major challenge for CRC immunotherapy. CD8
+ T cells play an essential role in tumour immunity, therefore, the promotion of functional
CD8
+ T cell infiltration to tumour microenvironment might improve the CRC therapy efficiency,
especially MSS type.
[0071] Microbiota contributes to antitumour efficiency by modulating host immune response,
working alone or facilitating immunotherapy. Microbiota depletion by antibiotic consumption
or germ-free mice was related with nonresponse to immunotherapy and oral supplementation
of bacteria or faecal microbial transplantation to antibiotic-treated or germ-free
mice restored the immunotherapy response. Though the necessity of microbiota in immunotherapy
was confirmed, whether and how some certain bacteria improve the antitumour efficiency
in CRC patients, especially in MSS-type are still not fully understood.
[0072] Gut microbiota affected cancer development by producing metabolites. Butyrate, as
one of the essential bacterial metabolites in colon, its concentration is negatively
correlated to CRC incidence. Butyrate producing bacteria including five
Roseburia OTUs were depleted, and Butyryl-coenzyme CoA transferase genes were reduced in the
microbiota of CRC patients. These data prompted us to investigate whether
R. intestinalis and its metabolite butyrate can contribute to the efficacy of antitumour immunity,
either alone or in combination with cancer therapy.
[0073] Roseburia intestinalis, a gram-positive, obligate anaerobic, butyrate-producing bacterium, firstly isolated
from human faecal material in 2002. Previously, we established a probe-based duplex
quantification PCR panel of bacteria biomarkers to diagnose colorectal cancer based
on two Asian cohorts.
R. intestinalis is one of the bacterial markers and depleted in CRC patients, but whether the altered
abundance of
R. intestinalis is a cause or consequence in CRC development needed further confirmation.
[0074] The protect role of
R. intestinalis in colorectal cancer was invested in two spontaneous AOM-induced and
ApcMin/+ murine models and
in vitro. R. intestinalis producing butyrate directly boosting cytotoxic CD8
+ T cells through Toll-like receptor 5 (TLR5) dependent NF-κB signalling.
R. intestinalis restricted MSI-type MC-38 and MSS-type CT-26 orthotopic tumour growth by modulating
antitumour CD8
+ T cell response, indicating that
R.
intestinalis and its metabolite butyrate can be supplied to improve the anti-colorectal cancer
therapy efficacy.
MATERIALS AND METHODS
Metagenomic cohorts of colorectal cancer patients and healthy controls
[0075] Four published metagenomic data cohorts from China, Germany, France, and Japan, including
334 CRC patients and 463 healthy individuals, were used to evaluate the abundance
of
R. intestinalis. Metagenomic data sets were downloaded from the European Nucleotide Archive (ENA)
with the following ENA identifiers: PRJEB10878 for YuJ_2017, PRJEB27928 for WirbelJ_2019,
ERP005534 for ZellerG_2014 and DNA Data Bank of Japan for YachidaS_2019 with accession
number: DRA006684 and DRA008156.
[0076] Another independent Chinese cohort named as "YuJ_2021" of 110 CRC patients and 112
healthy controls were recruited from the participants under colonoscopy screening
at the Jockey Club Bowl Cancer Education Centre, The Chinese University of Hong Kong.
The clinical study protocol was approved by Joint Chinese University of Hong Kong-New
Territories East Cluster Clinical Research Ethics Committee. All participants signed
an informed consent form. The mycobioa partial of the metagenomic data was published
by our team and the details of sample collection was shown in that study.
Metagenomic cohorts of responders and non-responders to immune checkpoint inhibitors
[0077] Metagenomic sequences data to analyze the relative abundance of
R. intestinalis in responders and non-responders to immune checkpoint inhibitors (ICI) were downloaded
from SRA under the Bioproject accession PRJNA751792 and PRJNA782662. This France cohort
included 333 non-small-cell lung cancer patients (258 non-responders and 75 responders).
Stool samples were collected before the ICI therapy start.
Targeted metabolomics cohort of colorectal cancer patients and negative controls
[0078] A published metabolomics cohort including 118 CRC patients and 128 negative control
individuals was performed gas chromatography coupled to time-of-flight mass spectrometer
(GC-TOFMS) targeted a human gut microbiota-host co-metabolism panel by the authors
and the significantly altered metabolites between CRC and NC were downloaded from
Pubmed. Metabolites with variable importance in projection (VIP) score >1 and P value
<0.05 were considered as significant.
Bacterial strain culture
[0079] Roseburia intestinalis (DSM 14610) was purchased from Leibniz Institute DSMZ-German Collection of Microorganisms
and Cell Cultures GmbH (Braunschweig, DE) and cultured at 37 °C in modified Yeast
Casitone Fatty Acids (YCFA) Broth with Hungate bottles filled with N
2.
E. coli MG1665 was used as a bacteria control and cultured at the same condition with
R. intestinalis. When the absorbance of
R. intestinalis reached to the optical density 600 (OD
600) of 1.0, the cultured medium was centrifuged and filtered with 0.22 µm pore size
filter to get the
R. intestinalis cultured medium (
R.i CM). We obtained the
E. coli cultured medium (
E. coli CM) at the same condition. Fractions larger than 3KD and less than 3KD were separated
with 3,000 Dalton molecular weight cutoff filters (UFC9003, Merck KGaA, Darmstadt,
DE).
Experimental animals
[0080] The intestinal adenoma spontaneous mouse strain C57BL/6J-
ApcMin/J (The Jackson Laboratory, Bar Harbor, US) was orally treated with 1×10
8 colony forming unit (CFU)
R. intestinalis daily at the age of 6 weeks, and they are raised for the following 6 weeks. Same
amount of
E. coli MG1665 was used as bacterial control and same volume of phosphate buffered saline
(PBS) was used as negative control.
[0081] 6 dosages of azoxymethane (AOM, A5486, Sigma-Aldrich, Darmstadt, DE) were injected
into 6-week-old C57BL/6 mice to induce sporadic tumour development. The same treatment
with the
ApcMin/+ model was used one week after the last dosage of AOM injection and lasted for 30
weeks continuously.
[0082] A MSI type mouse CRC cell line, MC-38 cell mix [5×10
5 cells in 10µL Matrigel (354248, Corning, Manassas, US) per mouse] was orthotopically
implanted into the rectum of C57BL/6. When the tumour was established or three days
after implantation, mice were started with
R.
intestinalis, E. coli or sodium butyrate (NaBu, 303410, Sigma-Aldrich, Darmstadt, DE) treatment and lasted
for three weeks. 1×10
8 CFU
R. intestinalis or
E. coli were given by oral gavage daily; 150 mM NaBu was provided in the drinking water and
refreshed every two days. Mice in other groups received saline in the same amount
of sodium as control. αPD-1 antibody (100µg per mouse, BP0146, Colone: RMP1-14, Bio
X Cell, West Lebanon, US) or control IgG (100µg per mouse, BP0089, Colone: 2A-3, Bio
X Cell) was injected intraperitoneally into MC-38 bearing mice, and followed by total
five injections with three days interval one week after MC38 implantation.
[0083] CT-26 tumour-bearing BALB/c mice with the same setting and treatment of MC-38 orthotopic
model was developed to evaluate the efficacy of αPD-1 antibody immunotherapy on microsatellite
stable (MSS)-type colorectal cancer. All experimental procedures were approved by
the Chinese University of Hong Kong Animal Ethics Committee.
Histology Evaluation
[0084] Formalin-fixed paraffin embedded colon tissues from each
ApcMin/+ and AOM-induced mouse were stained with hematoxylin and eosin (H & E) and sent to
pathologists for histology evaluation. Benign tissues were scored as "0"; tissues
with low grade dysplasia were scored as "1"; high grade dysplasia got "2".
Immunohistochemistry (IHC) Staining
[0085] To analyze the expression of Ki-67, ZO-1 and Claudin-3, colon tissue slides were
deparaffinized and rehydrated, followed with antigen unmasking and blocking. Slides
were then incubated with primary antibodies Ki-67 (D3B5) Rabbit mAb (12202, Cell Signalling
Technology, Danvers, US, 1:200), anti-ZO-1 antibody (ab96587, abcam, Cambridge, UK,
1:100) or Claudin-3 polyclonal antibody (34-1700, Thermo Fisher Scientific, Waltham,
US, 1:100) at 4 °C overnight, biotinylated secondary antibodies and streptavidin HRP
at room temperature for 30 minutes each. Signals were developed with DAB chromogen.
High-power fields were randomly shoot and the positive cells were calculated by ImageJ
1.53a with IHC Profiler plugin.
Fluorescein Isothiocyanate-Dextran intestinal permeability assay
[0086] Each mouse was orally gavaged with 150 µL of 80 mg/mL 4KDa FITC-dextran (68059, Sigma-Aldrich,
Darmstadt, DE) after 6 hours-fasting. About 50 µL blood was collected 4 hours after
FITC-dextran gavage. 100 µL of PBS diluted (1:5) plasma and standards were transferred
to a blank opaque-bottom 96-well plate and the fluorescence was determined at 528
nm with 485 nm excitation. Permeability was expressed as relative fluorescence units
between groups.
Western blotting analysis
[0087] To determine the expression of tight junction related proteins, total proteins from
mice colon tissues were extracted and separated by SDS-PAGE gel, then transferred
onto 0.2 µm Nitrocellulose Membrane (1620112, Bio-Rad, Redmond, US). The membrane
was blocked, incubated at 4°C overnight with primary antibodies anti-ZO1 tight junction
protein antibody (ab96587, abcam, Cambridge, UK, 1:500) and Claudin-3 polyclonal antibody
(34-1700, Thermo Fisher Scientific, Waltham, US, 1:1000), β-actin (13E5) Rabbit mAb
(4970, Cell Signalling Technology, Danvers, US, 1:1000) was used as internal control,
and followed with one-hour secondary antibody incubation at room temperature.
[0088] Primary antibodies Caspase-3 (8G10) Rabbit mAb (9665, 1:1000), Cleaved Caspase-3
(Asp175) antibody (9661, 1:1000), Caspase-7 antibody (9492, 1:1000), Cleaved Caspase-7
(Asp198) antibody (9491, 1:1000), Caspase-9 (C9) Mouse mAb (9508, 1:1000), PARP (46D11)
Rabbit mAb (9532, 1:1000), Cleaved PARP (Asp214) Rabbit mAb (5625, 1:1000), Cdk6 (D4S8S)
Rabbit mAb (13331, 1:1000), p27 Kip1 (D69C12) (3686, 1:1000) purchased from Cell Signalling
Technology (CST, Danvers, US) were used to analyse apoptosis and cell cycle related
protein expression on CRC cells HCT116, LoVo and SW480.
[0089] Phospho-NF-κB p65 (Ser536) Rabbit mAb (3033, Cell Signalling Technology, Danvers,
US, 1:1000) and NF-κB p65 (D14E12) Rabbit mAb (8242, Cell Signalling Technology, Danvers,
US, 1:1000) were used to measure NF-xB nuclear translocation in αCD3/αCD28/IL2 activated
Jurkat E6.1 T cells.
Cell culture
[0090] Normal human epithelial cell NCM460, human CRC cell lines HCT116, LoVo, SW480, and
mouse CRC cell lines MC38 and CT26 were cultured in Dulbecco's Modified Eagle's Medium
(DMEM) with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin (P/S). Jurkat
E6.1 (TIB-152, ATCC) was cultured in RPMI-1640 supplied with 10% FBS and 1% penicillin-streptomycin
and activated in 3 µg/ml anti-CD3 (300332, BioLgend, San Diego, US), 5 µg/ml anti-human
CD28 (302934, BioLgend, San Diego, US) and 100 IU/ml recombinant human IL-2 (589104,
BioLgend, San Diego, US) pre-coated wells for 3 days.
Cell viability assay and colony formation assay
[0091] 1000 cells per well were seed into a 96-well plate and treated with alive or pasteurized
E. coli (CFU=10
6/ml) and
R. intestinalis (CFU=10
6/ml), 5% (vol/vol) YCFA,
E. coli CM,
R. i CM in DMEM separately. Cell viability was determined by Thiazolyl Blue Tetrazolium
Bromide (M5644, Merck KGaA, Daemstadt, DE) 3 days constantly.
[0092] For cell colony formation assay, same amounts of cells were seed into 6-well plates,
followed with the same treatments as cell viability assay for one to two weeks and
changed the culture medium freshly every 3 days. After cold Methanol fixation, cells
were stained with 0.5% crystal violet solution. Cell colonies were measured with ImageJ
1.53a.
Immunocytochemistry (ICC) staining
[0093] Human CRC cells seed on poly-L-lysine coated coverslips in 4-well plate and fixed
with ice-cold methanol for 10 minutes. Then, cells were blocked with 1% BSA in PBST
for 30 minutes and incubated with Ki-67 (8D5) mAb (9449, Cell Signalling Technology,
Danvers, US, 1:6400) in a humidified chamber at 4°C overnight and Alexa Fluor 488
goat anti-mouse IgG secondary antibody (A-11001, Thermo Fisher Scientific, Waltham,
US, 1µg/mL) for one hour at room temperature in the dark. Cell nuclear was stained
with DAPI (P36935, Thermo Fisher Scientific, Waltham, US) for one minute for cell
counting before mounting. Images were captured with a confocal laser scanning microscope
(TCS SP8, Leica, Buffalo Grove, US). Ki-67 positive cell percentage was Alexa Fluor
488 positive cell amounts in total cell amounts.
Cell apoptosis and cell cycle
[0094] Flow Cytometry was used for cell apoptosis and cell cycle analysis with FACSAria
cell sorter (BD Biosciences, Franklin Lakes, US). Data was analyzed by FlowJo 10.4.
[0095] Cell apoptosis was detected by FITC Annexin V apoptosis detection kit (556547, BD
Biosciences, Franklin Lakes, US). Adherent cells were removed by Trypsin-EDTA solution
and washed with cold PBS twice and then resuspend in binding buffer at a concentration
of 1 × 10
6 cells/ml. 100 µl of cell solution was transferred to a 5ml tube, incubated with 5
µl FITC Annexin V and 5 µl PI for 15 minutes at 25 °C in the dark. Cells were added
with 400 µl binding buffer to stop staining and analyzed by flow cytometer in one
hour.
[0096] For cell cycle analysis, cell pellet was washed by PBS and incubated in 70 to 80%
ethanol at -20°C for more than two hours. In each test, 10
6 cells were used for staining in 0.5 ml PI/RNase staining buffer (550825, BD Biosciences,
Franklin Lakes, US) for 15 minutes at room temperature in the dark. Tubes were stored
at 4 °C protected from light and analyzed on flow cytometer with one hour.
Patient-derived colorectal cancer organoid culture
[0097] CRC patient-derived organoid was obtained from the Princess Margaret Living Biobank
(PMLB), originally from a 46-year-old female colorectal adenocarcinoma patient. The
minced pathologic specimens were digested and embedded into Matrigel and maintained
in Advanced DMEM/F12, supplied with 1% P/S, 10 mM HEPES, 1×GlutaMAX medium containing
N2 and B27 supplements, 50% (vol/vol) Wnt3a conditional medium, 10% (vol/vol) R-spondin-1
conditional medium, 100ng/ml Noggin conditional medium, 50ng/ml EGF, 10nM Gastrin,
100ng/ml FGF10, 1.25 mMN-acetylcysteine, 500nM A8301, 1µM SB202190, 10mM Nicotinamide.
Treatments containing 5% (vol/vol) YCFA,
E. coli CM or
R. i CM were added into the culture medium directly and freshly changed every three days.
Surface area of organoid was measured by Image J 1.53a.
Untargeted metabolomic profiling
[0098] Metabolomic profiling was performed by BIOTREE, Shanghai, China. 400µL extract solution
containing isotopically labelled internal standard mixture was added into100µL each
sample of YCFA<3kDa,
E. coli CM<3kDa and
R.i CM<3kDa, sonicated in iced water bath for 10 minutes, and incubated at -40 °C for
one hour to precipitate proteins. After centrifuge at 13800 g for 15 minutes at 4
°C, the supernatant was transferred to a new glass vial for liquid chromatography
tandem mass spectrometry (LC-MS/MS) analysis using an ultra-high performance liquid
chromatography (UHPLC) system (Vanquish, Thermo Fisher Scientific, Waltham, US) with
a UPLC BEH Amide column (2.1 mm × 100 mm, 1.7 µm), coupled to Orbitrap Exploris 120
mass spectrometer (Orbitrap MS, Thermo Fisher Scientific) on information-dependent
acquisition (IDA) mode controlling by the acquisition software (Xcalibur, Thermo Fisher
Scientific). Electrospray ionization (ESI) source conditions were as following: sheath
gas flow rate was 50 Arb, Aux gas flow rate was 15 Arb, capillary temperature was
320 °C, full MS resolution was 60000, MS/MS resolution was15000, collision energy
was 10/30/60 in NCE mode, spray voltage was 3.8 kV (positive) or -3.4 kV (negative)
respectively.
Targeted short chain fatty acid gas chromatography-mass spectrometry analysis
[0099] To confirm the findings from untargeted metabolomic profiling, metabolites were extracted
from less than 3 kDa fractions of YCFA,
E. coli CM and
R.i CM for analysis with gas chromatography-mass spectrometry (GC-MS, GC2030-QP2020 NX,
Shimadzu, Tokyo, JP) utilized with an Agilent HP-FFAP capillary column (30m×250µm×0.25µm,
J&W Scientific, Folsom, US). 1 µL aliquot of sample was injected in split mode (5:1).
Helium was used as the carrier gas. Front inlet septum purge flow was 3 ml/min, and
column flow was 1 ml/min. The program of oven temperature ramp was as follow: 80 °C
hold on 1 min; then raised to 200 °C at a rate of 10 °C/min
, hold on 5 min; then raise to 240 °C at a rate of 40 °C/min and hold on 1 min. The
front injection, transfer line, ion source and quad temperatures were 240 °C, 240
°C, 200°C and 150 °C. The electron energy was -70 eV. The mass spectrometry data were
acquired in Scan/SIM mode with the m/z range of 33-150 after a solvent delay of 3.5
min. Standard curves were established with acetic acid (64-19-7, Dr. Ehrenstorfer,
Augsburg, DE), propionic acid (79-09-4, Dr. Ehrenstorfer, Augsburg, DE), isobutyric
acid (79-31-2, Dr. Ehrenstorfer, Augsburg, DE), butyric acid (107-92-6, Dr. Ehrenstorfer,
Augsburg, DE), isovaleric acid (503-47-2, Dr. Ehrenstorfer, Augsburg, DE), valeric
acid (109-52-4, Dr. Ehrenstorfer, Augsburg, DE), hexanoic acid (142-62-1, Dr. Ehrenstorfer,
Augsburg, DE), heptanoic acid (111-14-8, Dr. Ehrenstorfer, Augsburg, DE), octanoic
acid (124-07-2, Dr. Ehrenstorfer, Augsburg, DE), nonanoic acid (112-05-0, Dr. Ehrenstorfer,
Augsburg, DE), decanoic acid (334-48-5, Dr. Ehrenstorfer, Augsburg, DE), 2-Methylvaleric
acid (97-61-0, Dr. Ehrenstorfer, Augsburg, DE).
T cell isolation and activation
[0100] T cells isolated from healthy human peripheral blood mononuclear cells (PBMC) using
immunomagnetic negative selection kit (17951, Stemcell Technologies, Vancouver, CA).
Isolated T cells were then cultured in complete RPMI-1640 medium supplied with human
CD3/CD28 magnet beads (11161D, Thermo Fisher Scientific, Waltham, US) and 30 U/ml
recombinant human IL-2 (589104, BioLgend, San Diego, US) for expiation and activation
for three days. Mouse T cells were isolated from the spleen of naive mice and purified
with Mouse T cell isolation kit (19851, Thermo Fisher Scientific, Waltham, US), cultured
in complete RPMI-1640 medium with mouse CD3/CD28 beads (11456D, Thermo Fisher Scientific,
Waltham, US) and 30 U/ml recombinant mouse IL-2 (575404, BioLegend, San Diego, US).
Immune cell characterization
[0101] Cytotoxic CD8
+ T cell markers Granzyme B, IFN-γ, TNF-α under the
in vitro treatments of
R.i CM, NaBu or TLR5 inhibitor TH1020 (HY-116961, MedChemExpress, Monmouth Junction,
US) were analyzed with flow cytometry. Isolated human T cells were stimulated with
30ng/ml PMA, 1µg/ml ionomycin and 2.5 µg/ml monensin in complete RPMI-1640 medium
at 37 °C for 4 hours. For surface antigens staining, cells were stained with Brilliant
Violet 605 anti-human CD45 antibody (2D1) (368524, BioLegend, San Diego, US), PE anti-human
CD3 antibody (OKT3) (317308, BioLegend), PE/Cyanine5 anti-human CD4 antibody (RPA-T4)
(300510, BioLegend), Brilliant Violet 421 anti-human CD8a antibody (SK1) (344748,
BioLegend) in FACS buffer on ice for 30 minutes. After fixing with Transcription Factor
Fixation/Permeabilization buffer (00-5521-00, Invitrogen, Waltham, US), intracellular
cytokines were stained in 1 × Permeabilization buffer (00-8333-56, Invitrogen, Waltham,
US) with PE/Cyanine7 anti-human/mouse Granzyme B recombinant antibody (QA16A02) (372214,
BioLegend), Brilliant Violet 711 anti-human IFN-γ antibody (4S.B3) (502540, BioLegend),
FITC anti-human TNF-α antibody (Mab11) (502906, BioLegend). Granzyme B, IFN-γ, TNF-α
negative cells were gated in CD8
+ T cells (CD45
+ CD3
+ CD8
+) without intracellular staining.
[0102] For tumour-infiltrating immune cell analysis, tumour from MC-38 and CT-26 bearing
rectal orthotopic mouse models was minced on ice and digested in PBS buffer containing
0.5mg/mL collagenase IV and 0.25mg/ml DNase I at 37 °C for 30 min. After digestion,
tumour tissue was filtered through a 70 µm cell strainers, and then distributed into
different panels. For the surface staining of MDSC, cells were stained with Brilliant
Violet 605 anti-mouse CD45 antibody (30-F11) (103140, BioLegend), PerCP/Cyanine5.5
anti-mouse/human CD11b antibody (M1/70) (101228, BioLegend), PE anti-mouse Ly-6G/Ly-6C
(Gr-1) antibody (RB6-8C5). MDSC was gated in CD45
+ CD11b
+ Gr-1
+. For CD8 functional markers staining, the procedures were similar with the
in vitro T cell analysis. Antibodies used for staining are as following: Brilliant Violet
605 anti-mouse CD45 antibody (30-F11) (103140, BioLegend), PE anti-mouse CD3 antibody
(17A2) (100206, BioLegend), Brilliant Violet anti-mouse CD8a antibody (53-6.7) (100738,
BioLegend), PE/Cyanine5 anti-mouse CD4 antibody (GK1.5) (100410, BioLegend), PE/Cyanine7
anti-human/mouse Granzyme B recombinant antibody (QA16A02) (372214, BioLegend), FITC
anti-mouse IFN-γ antibody (XMG1.2) (505806, BioLegend), Brilliant Violet 711 anti-moue
TNF-α (MP6-XT22) (506349, BioLegend). Flow Cytometry was performed on FACSAria cell
sorter (BD Biosciences, Franklin Lakes, US). Data was analyzed by FlowJo 10.4.
RNA isolation and quantitative real-time PCR (qPCR)
[0103] Total RNA was extracted with Trizol (15596018, Invitrogen, Waltham, US) from naive
mouse spleen isolated T cells after CD3/CD28/IL-2 activation and
R.i CM and Butyrate treatments. Then, RNA was reverse transcript into cDNA with RT reagent
kit with gDNA eraser (RR047A, Takara, Tokyo, JPN). qPCR was performed using TB Green
Premix (RR420A, Takara) in the QuantStudio 7 Flex System (Thermo Fisher Scientific,
Waltham, US). The relative RNA expression was normalized to that of
β-actin as denominators.
RESULTS
Roseburia intestinalis is depleted in stool samples of colorectal cancer patients
[0104] Besides the cohorts to discover the novel noninvasive CRC diagnosis method in our
previous study, the abundance of
R. intestinalis was evaluated in four other published metagenomic data cohorts from China, Germany,
France, and Japan, including 334 CRC patients and 463 healthy individuals as well
(
Figure 8A). Another inhouse Chinese cohort of 110 CRC patients and 112 healthy controls were
also recruited from the participants under colonoscopy screening to investigate the
R. intestinalis abundance (
Figure 8B). In these five cohorts,
R. intestinalis was depleted in the faecal samples of CRC patients.
[0105] To link the gut microbiota-associated metabolites to colorectal carcinogenesis, metabolomic
profiling was performed on faecal samples from 118 CRC patients and normal controls,
and butyric acid was depleted in CRC patients (
Figure 8C).
R. intestinalis was also depleted in CRC patients in this study. The negative associations of
R. intestinalis and butyrate with CRC from human faecal metagenomic and metabolomic sequencing data
indicate their potential tumour suppressive role on CRC.
[0106] After reanalysis of a published metagenomics cohort, we found that
R. intestinalis was enriched in the stool samples of 75 immune checkpoint inhibitor responders compared
to 258 non-responders in non-small-cell lung cancer (
Figure 8D), which could be used as a biomarker to refine patient stratification to immunotherapy.
Roseburia intestinalis inhibited colorectal cancer development in murine colorectal cancer models
[0107] Two spontaneous mouse models were used to evaluate the tumour suppressive role of
R. intestinalis on CRC. We started daily gavaging 6-week-old
ApcMin/+ mice R. intestinalis (
R.i) or
E. coli strain MG1655 for continuous 6 weeks (
Figure 9A).
R. i significantly reduced the colon tumour incidence (
Figure 9B, 9C), tumour number and size (
Figure 9D) compared those treated with
E. coli and PBS. Besides, colon tissues from
R. i treated mice got lower dysplasia score under histology evaluation and obviously less
Ki-67 positive cell proportion was observed in both tumour and non-tumour colon tissues
(
Figure 9E). The mouse body weight, liver (ALT, AST) and renal (BUN, CREA) function were not
obviously altered with the dosage and frequency of
R.i treatment (
Figure 10A and 10B). We got consistent results in another carcinogen-induced CRC model in which 6-week-old
C57BL/6J mice were injected with 6 dosages AOM weekly, following with 30 weeks administration
of
R. intestinalis (
Figure 9F-K). What's more,
R. intestinalis enhanced gut barrier homeostasis (
Figure 11). Lower serum FITC-dextran (4kd) concentration was detected four hours after its
orally gavage, indicating that barrier impairment was milder in
R.i treated mice (
Figure 11A). Molecularly, tight junction protein ZO-1 and Cluaindin-1 expression lever was significantly
higher with
R.i treatment (
Figure 11B, 11C). Taking together,
R. intestinalis protected mice against colorectal carcinogenesis by deducing tumour incidence, decreasing
tumour number and tumour size, and enhancing gut barrier functions under a well-tolerated
dosage.
Roseburia intestinalis cultured medium induces colorectal cancer cell apoptosis and cell cycle arrest
[0108] Based on the findings in murine models, we then detected whether
R. intestinalis itself or its producing metabolites have the antitumour effects in vitro. First,
colon cancer cell lines HCT116, LoVo, SW480 and normal epithelial cell line NCM460
were cocultured with alive or pasteurized
E. coli or
R.i for 4 hours per day for continuous 3 days, but we didn't observe any difference in
cell viability among groups, indicating that
R.
intestinalis itself has no effects on colon cell viability with this coculture condition (
Figure 12).
[0109] We then moved to the metabolites produced by
R. intestinalis. The cell viability of HCT116, LoVo and SW480, but not NCM460 was inhibited after
5% (vol/vol)
R. intestinalis cultured medium (
R.i CM) treatment (
Figure 13A). The results were confirmed by colony formation assay (
Figure 13B) and nuclear proliferating marker Ki-67 immunocytochemistry staining (
Figure 13C). Moreover,
R.i CM induced higher apoptosis cell proportion at both early and late stage in CRC cell
lines (
Figure 13D). Apoptosis markers Cleaved Caspase-7 and Cleaved PARP increased in all three CRC
cells, while Cleaved Caspase-3 and Cleaved Caspase-9 increased in HCT116 and SW480
at protein level after
R.i CM treatment (
Figure 13E). In addition, HCT116, LoVo and SW480 treated with
R.i CM showed a cell cycle arrest with an increased cell proportion at GO/G1 phase and
a decrease of cells in synthesis (S) (
Figure 13F). The protein level of cell cycle marker Cdk6 was increased and p27 Kip1 was increased
after treatment with
R.i CM, indicating that
R.i CM induces G1 phase arrest (
Figure 13G). Besides of cell lines, CRC patient derived organoid was also treated with
R.i CM and the surface area of it was extremely lower after the treatment (
Figure 13H). The results indicate that metabolites producing by
R. intestinalis mediated the tumour suppressive effects
in vitro.
Butyrate is the main tumour suppressive component produced by Roseburia intestinalis
[0110] To determine the effective components in
R.i CM, we separated it into fractions less than 3 kDa and more than 3 kDa. Only
R. i CM<3 kDa still kept the tumour-suppressive effects on Colon cancer cells, but not
R.i CM>3 kDa (
Figure 14A).
R.i CM<3 kDa was sent for untargeted metabolomics profiling by LC-MS/MS and butyric acid
ranked top ones (
Figure 14B). The accurate concentration of short chain fatty acid
was measured by targeted gas chromatography-mass spectrometry (GC-MS), and butyric acid
was markedly enriched in R.i CM<3 kDa fractions (
Figure 14C). The concentration of butyric acid in
R.i CM reached to more than 2000 µg/ml (
Figure 14C). The elevation of butyrate acid was confirmed in
ApcMin/+ and AOM-induced mouse faecal materials by GC-MS analysis (
Figure 14D).
[0111] The direct suppressive effect of butyrate on colorectal cancer cells was confirmed
by cell viability inhibition (
Figure 15A), apoptosis inducement (
Figure 15B) and cell cycle arrest (
Figure 15C) with the treatment of comparable concentration (1mM) of butyrate acid in 5%
R.i CM.
[0112] In another set of untargeted metabolomics profiling by GC-MS, inosine was the most
enriched molecular in
R.i CM>3 kDa (
Figure 16A), indicating that inosine might be another tumour suppressive component in
R.i CM and the concentration of inosine in
R.i CM is around 8µg/ml (
Figure 16B). But comparable dosage of inosine in
R.i CM has no anti-tumour effects in vitro (
Figure 17A-C).
Roseburia intestinalis cultured medium and butyrate directly boosts cytotoxic CD8+ T cells in vitro
[0113] Besides the direct apoptosis inducement and proliferation inhibition of butyrate
and
R. intestinalis metabolites on tumour cells, we also estimated their antitumour response on CD8
+ T cells, which play a central role in tumour immunity. T cells were isolated from
human
peripheral blood mononuclear cell or mouse spleen and activated by α-CD3/CD28 and IL-2 with or without
R.i CM or butyrate (1mM) treatment. The results show that both
R.i CM and butyrate treatment promoted the production of Granzyme B, IFN-γ and TNF-α
in CD8
+ T cells (
Figure 15D, Figure 18). To understand how
R. intestinalis facilitates CD8
+ T cells by producing butyrate, the mRNA expression level of receptors and transporters
on CD8+ T cells, including
Gpr41, Gpr43, Gpr109a, Slcl6a1, Tlr1 to 6, Id2, Il2ra, Il2rb, Il7r, Il12rb1, Il12rb2,
Cd8a, Cd8b1, Cd27, Cd28, Ox40, 4-lbb, were measured by qPCR with or without
R.i CM treatment (
Figure 15E). Unexpectedly,
R. i CM treatment didn't change the expression of short chain fatty acid receptors or
transporters, but other effector T cell-associated genes, like
Id2, Il2ra and
Cd28, and pattern recognition receptor
Tlr5 (
Figure 15E). Especially, the
Tlr5 mRNA expression level was more than 10 times higher with
R.i CM treatment (
Figure 15E). Similarly,
Tlr5 expression was much higher in CD8
+ T cells treated with butyrate (
Figure 15F). These data led us to hypothesize that
R. intestinalis may facilitate CD8
+ T cell response on a TLRS-dependent pathway. TLR5 captured on Chip CMS can bind butyric
acid with an affinity of 264 µM as determined by surface plasmon resonance (SPR) (
Figure 15G). To confirm our hypothesis, a selective TLRS antagonist TH1020 was used to inhibit
TLR5 signalling. Obviously, Th1020 abolished the effective promotion of butyrate on
Granzyme B, IFN-γ and TNF-α in CD8
+ T cells (
Figure 15H). Molecularly, both
R.i CM and butyrate increased NF-κB nuclear translocation with higher phospho-NF-xB expression
and inhibited by Th1020 in α-CD3/CD28/IL-2 activated Jurkat E6.1 T cells (
Figure 15I). Overall, butyrate, produced by
R. intestinalis, boosted cytotoxic CD8
+ T cells through TLR5-NF-κB activation.
[0114] The effect of comparable dosage of inosine (0.4µg/ml) on promotion of Granzyme B,
IFN-γ and TNF-α in CD8
+ T cells was also detected (
Figure 17D).
Supplementation of Roseburia intestinalis and butyrate restricts MC38 and CT-26 orthotopic tumour growth
[0115] We next tested whether
R. intestinalis and butyrate could effectively enhance immunotherapy on colorectal cancer. Treatments
of
E. coli, R.i or sodium butyrate (NaBu) started three days after murine CRC cell MC-38 injection
into C57BL/6J mouse's rectum and lasted for three weeks, αPD-1 therapy began after
the tumour was established (
Figure 19A). R. intestinalis, NaBu and αPD-1 treatments inhibited MC38 orthotopic tumour growth (
Figure 19B-D). Combination of αPD-1 with
R.i or NaBu did not enhance the efficiency of αPD-1 therapy; while tumours from
R.i or NaBu-treated mice are small already (
Figure 19B-D). R.
intestinalis or sodium butyrate treatment of MC38 tumours increased infiltrating CD8
+ T cells (
Figure 19E) and Granzyme B
+, IFN-γ
+ and TNF-α
+ CD8+ T cells (
Figure 19F) while decreasing MDSCs (
Figure 19G). In this αPD-1 sensitive mouse model,
R. intestinalis and butyrate supplementation is as effective as αPD-1 therapy.
[0116] In another MSS-type mouse colorectal tumour model, CT-26 was injected to BALB/c mouse
rectum under same experimental design and timeline with last model (
Figure 20A). CT-26 was non-sensitive to αPD-1 therapy, but the tumour weight and size are still
reduced with
R.i or NaBu treatment alone compared with αPD-1 therapy and control groups (
Figure 20B-D). Moreover, the combination of
R. intestinalis or sodium butyrate with anti-PD-1 further heightened the anti-tumour effects of
R. intestinalis or sodium butyrate (
Figure 20B-D). Consistently, the proportion of infiltrating CD8+ T cells (
Figure 20E) and Granzyme B
+, IFN-γ and TNF-α
+ CD8
+ T cells was significantly increased in CT26 tumours with monotreatment of
R. intestinalis or sodium butyrate (
Figure 20F), and their combination with anti-PD-1 showed additional effects on these immune
cells (
Figure 20E-F). Treatment of
R. intestinalis or sodium butyrate but not anti-PD-1 monotherapy also reduced the proportion of MDSCs
in tumours (
Figure 20G).
R. intestinalis and sodium butyrate are effective on facilizing anti-PD-1 therapeutic efficacy against
CRC with MSS phenotype.
DISCUSSION
[0117] Faecal butyrate was found closely related to CRC incidence in the inventors' previous
study, indicating the potential therapeutic role of butyrate. Detected by LC-MS/MS,
the inventors found that the anticancer efficiency from
R. intestinalis was mainly from butyrate. They then confirmed that the cultured medium of
R. intestinalis restricted the proliferation of CRC cells but not normal epithelial cells by apoptosis
induction and cell cycle arrest. Supplement of sodium butyrate to mice achieved the
same anti-tumour response. Butyrate in lumen is the primary energy source for normal
colonocytes. Due to the Warburg effect, tumour cells rely on glucose as their primary
energy, which lead to the accumulation of butyrate and act as histone deacetylase
(HDAC) inhibitor. The Warburg effect dictates the mechanism of butyrate mediated histone
acetylation and inhibition of tumour cell proliferation but not normal cells. Directly,
by producing butyrate,
R. intestinalis kills tumour cells to inhibit the progression of CRC.
[0118] The promotion role of butyrate producing bacteria
R. intestinalis on immune checkpoint inhibitor response was confirmed by reanalysing a published
metagenomics cohort (
Figure 8D). Other butyrate producing bacteria such as
Akkermansia muciniphila and
Faecalibacterium were also reduced in PD-1 therapy non-responders. Moreover, faecal butyric acid concentration
is associated with PD-1 inhibitor efficacy in solid cancer. These association analyses
between butyrate concentration or butyrate producing bacteria abundance and immunotherapy
response inspired us the therapeutic potential of butyrate on CRC.
[0119] Clinically, the major challenge of immunotherapy on CRC patients is the poor response,
especially for most patients presenting MSS CRC tumours. Even in MSI-H CRC patients,
only 40% of them responded to immune checkpoint inhibitors. In practical mouse experiments,
MSS type CT-26 cells are considered as sensitive to immune checkpoint blocking therapy.
As illustrated in the results, anti-PD-1 therapies were effective in MC-38 bearing
mouse (
Figure 19), but not CT26 bearing mouse (
Figure 20). In MC-38 rectum orthotopic mice, both
R. intestinalis and butyrate were as efficient as anti-PD-1 on tumour shrink.
R.
intestinalis and butyrate alone were sufficient to suppress the tumour growth without any additional
improvements when combining with anti-PD-1. In CT26 rectum orthotopic mice, the anti-tumour
immunity of
R. intestinalis and butyrate were more obvious when anti-PD-1 therapy was disabled in this model.
Thus,
R. intestinalis provides a promising therapy in CRC patients who are resistant to the current anti-PD-1
therapy.
[0120] With a focus on the specific immune cell type, the current immunotherapies are centred
on CD8
+ T cells, which are the most powerful anticancer immune effectors by recognizing and
killing tumour cells with the cytotoxic molecules. However, the direct role of microbiota
on the CD8
+ T cells functional regulation needs further study. Different from other existing
studies, in which bacteria only served as enhancers of immunotherapy, the inventors
found that
R. intestinalis itself could promote anti-tumour cytotoxic CD8
+ T cell response directly through its metabolite butyrate, independently from anti-PD1
therapy, especially in MSS-type CT-26 bearing mice.
[0121] Molecularly downstream, short chain fatty acids (SCFAs) interact with metabolite-sensing
G protein-coupled receptors GPR 41, GPR 43 and GPR109A on epithelial cells. But in
the inventors' study, stimulation of butyrate didn't change the mRNA expression of
Gpr 41, Gpr 43 and
Gpr109a, but
Tlr5 on CD8
+ T cells. In another study, treating isolated CD8
+ T cells from GPR43 or GPR109a KO mice with butyrate still induced cytotoxic CD8
+ T cells and blocking GPR-signaling with pertussis toxin did not impair the effect
of butyrate on functional CD8
+ T cells, which indicating that the promotion of butyrate on CD8
+ T cell function may not depend on GPR receptors. Butyrate increased the binding of
the transcription factor specificity protein 3 to the TLR5 promotor regions to upregulate
the transcription of TLR5, which confirm the association of butyrate with TLRS at
the transcription level indirectly, but whether butyrate directly binding to TLRS
is unknown. The inventors confirmed the binding affinity of butyric acid to TLRS by
surface plasmon resonance (SPR), indicating that TLR5 might be a butyrate receptor,
at least on CD8
+ T cells. TLR5, as one of the pathogen-associated molecular patterns (PAMPs), expressed
in epithelial cells and immune cells, specifically recognize flagellin on bacteria.
Activation of TLRS translocated the nuclear factor NF-κB and stimulated the production
of TNF-α. TH1020, identified as an antagonist of TLR5, competed with flagellin and
disrupted the association with TLRS based on molecular docking simulation, repressed
the expression of downstream TNF-α signaling pathways. In this study, the effects
of butyrate on CD8
+ cells were abolished by TH1020, with the repression of Granzyme B, IFN-γ, TNF-α and
NF-κB mobilization. Overall, butyrate directly boosting cytotoxic CD8
+ T cells through TLRS stimulation and NF-xB signalling activation.
[0122] The inventors thus discovered in human faecal sequencing data,
R. intestinalis and butyrate were depleted in CRC patients compared to healthy controls. By using
multiple mouse models, they demonstrated the preclinical efficiency of
R. intestinalis supplements as monotherapy on restricting colorectal tumour growth through butyrate,
especially on MSS-type CT-26 tumours. Molecularly,
R. intestinalis cultured medium and butyrate suppressed CRC by directly inducing tumour cell apoptosis,
arresting cell cycle, and mediating cytotoxic Granzyme B
+, IFN-γ
+, TNF-α
+ CD8
+ T cells through TLRS dependent NF-xB signalling.
R.
intestinalis, as a promising probiotics supplement, opened a new way to expand the CRC treatment
efficiency, particularly for immunotherapy resistant patients.
EXAMPLE 3: Streptococcus salivarius k12 Reprograms Tumour Immune Microenvironment to Synergize with Anti-PD1 to Suppress
Colorectal Tumourigenesis
BACKGROUND OF THE INVENTION
[0123] Colorectal cancer is one of the leading cancer deaths worldwide. Immune checkpoint
inhibitors (ICI), especially, monoclonal antibodies (mAbs) directed at the PD-1/PD-L1
(programmed cell death protein-1/programmed death ligand-1) axis has revolutionized
cancer treatment. However, only a minority of patients respond to these immune-oncology
therapies, and undeniably, a multitude of ICI-associated immune-related adverse effects
(irAEs) remain a clinical challenge. Identification of optimized therapeutic analogous,
immunostimulatory molecules that trigger activations of innate and adaptive immune
responses to facilitate the successful clinical immunotherapy, is therefore highly
warranted.
[0124] In recent years, the "favorable" and "unfavorable" gut microbiota has been highlighted
in the immune-oncology study. Several studies have pointed out the involvement of
some specific probiotics in modulating the response to immunotherapy. Of note, administration
of
Bifidobacterium longum and
Bifidobacterium breve cocktail to TAC mice is sufficient to control the tumour growth by increasing anticancer
T cells to the same extent as anti-PD-L1 through activating dendritic cell-mediated
immune responses, whereas the synergistic effect from combined
Bifidobacterium spp with PD-1/PD-L1 blockade can almost abolish the tumour growth.
[0125] Using shotgun metagenomic sequencing, the inventors identified
S. salivarius as the most significantly depleted probiotic in the stool samples of patients with
CRC, indicating its possible role in preventing colorectal tumourigenesis. As there
has been no reported functional role
of S. salivarius (M18) in suppressing CRC, and due to the strain-specific efficacy of the probiotics,
the inventors further evaluated the protective role of
S. salivarius (K12) and a healthy human isolated strain on colorectal tumourigenesis. They found
the specific tumour suppressive effect of
S.
salivarius (K12) as compared with
S. salivarius (M18) and
S. salivarius (Human). Through bacterial whole genome sequencing (WGS), the inventors found the
S.
salivarius (K12) contains a specific gene cluster responsible for exopolysaccharides (EPS) production.
They also revealed that
S. salivarius (K12) can augment the intratumoural effector CD8+ T cells infiltration, which turns
the "cold" tumours into "hot" tumours to potentiate the anti-PD1 response in CRC.
MATERIALS AND METHODS
Animal experiments
[0126] Animal models were established the same as described previously. In brief, C57BL/6J
-ApcMin/J mice, which develop intestinal polyps spontaneously were used as a model of spontaneous
intestinal neoplasia. They were purchased from the Jackson Laboratory (Bar Harbor,
ME, USA) and maintained in the animal facility at the Chinese University of Hong Kong.
Mice at 4-5 weeks old were divided into 3 groups - 1×10
8 colony forming units (CFU)
of S. salivarius or
E. coli MG1655, or the same volume of BHI was gavaged to them once daily for 10 weeks.
Faecalibacterium prausnitzii, which was reported to have an immune modulatory effect, was used as a positive control.
Colorectal tumour formation was monitored by mouse colonoscopy (Coloview, Karl Stroz,
Germany). Colonic tissues were collected for immune profiling.
[0127] For the carcinogen-induced CRC mouse model, male C57BL/6 mice at 6 weeks old were
intraperitoneally injected with a single dose of 10mg/kg AOM (Merck, Darmstadt, Germany),
followed by 2% DSS (MP Biomedicals, Solon, OH) administration for 1 week. After DSS
treatment, the same treatment regimen with the
Apcmin/+ mice was used in this AOM/DSS-induced CRC model. Mice were raised to 20 weeks for
the evaluation of the probiotic treatment efficacy.
[0128] For the xenograft model, male C57BL/6 mice or BALB/c mic at 5 weeks old were pre-treated
with 1×10
8 CFU
S. salivarius. E. coli MG1655, or the same volume of BHI for 2 weeks, following which, the mice were subcutaneously
injected with MC38 (5×10
5 cells) or CT26 (1×10
6 cells) into the flank, respectively. Tumour volume was measured with a digital caliper
every other day after the outgrowth of the tumour. The anti-mouse programmed cell
death protein 1 (anti-PD1) monoclonal antibody (BE0146, Bio X Cell, Lebanon, NH) or
IgG isotype control (BE0089, Bio X Cell) was administrated to the CT26 syngeneic mice
to evaluate the synergistic effect
of S. salivarius in improving anti-PD1 response. At the end of the experiment, tumour tissues were
collected for evaluating the changes of the tumour immune prefile. All the procedures
were performed in accordance with guidelines approved by the Animal Experimentation
Ethics Committee of The Chinese University of Hong Kong.
Bacterial strains and culture conditions
[0129] S. salivarius (M18 and K12) were purchased from American Type Culture Collection (ATCC; Manassas,
VA).
E. coli MG1655 was obtained from DSMZ. They were cultured in BHI broth (Thermo Fisher Scientific,
West Palm Beach, FL) at 37°C under aerobic conditions. The bacteria were centrifuged
at 5,000 rpm for 10 minutes to obtain the bacteria pallet and resuspended in BHI before
gavaging to mice. The
S. salivarius conditioned medium (Ss. CM) was obtained by centrifuging the bacteria culture supernatant
at 4500g for 15 mins and filtered through a 0.2-µm pore-size filter.
[0130] For the
S. salivarius (human isolation), healthy human faecal samples were collected and suspended in PBS
within 30 minutes. After homogenization, the suspension was filtered through a 100µm
strainer to remove larger particles. Serial dilution was then performed by using the
faecal suspension and the BHI agar plate was further used to incubate the suspension
in an aerobic incubator at 37 °C for 24 hours. Colonies with a similar appearance
to
S.
salivarius were purified by streak plated method. The 16S rDNA for each isolate was amplified
by PCR with the university primers 341F (5'- CCTAYGGGRBGCASCAG-3', SEQ ID NO: 1) and
806R (5'-GGACTACNNGGGTATCTAAT-3', SEQ ID NO:2).
S.
salivarius (human) was identified by blasting the 16S rDNA in the GenBank.
Cell culture
[0131] Colon cancer cell lines, including HCT116, Lovo, MC38, and CT26, were obtained from
American Type Culture Collection (ATCC). Normal colonic epithelial cell line NCM460
was obtained from INCELL Corporation (San Antonio, TX). All the cell lines were cultured
in high-glucose Dulbecco's Modified Eagle's Medium (DMEM) (Thermo Fisher Scientific)
supplemented with 10% (vol/vol) fetal bovine serum (FBS) (Thermo Fisher Scientific),
1% penicillin/ streptomycin in a humidified atmosphere containing 5% CO
2. For co-culture of epithelial cells with bacteria, the penicillin/streptomycin in
the culture medium was removed and cells were exposed to bacteria with a multiplicity
of infection (MOI) of 200 for 2 hours. Cell viability was determined by 3-(4,5-dimethylthiazoly-2-yl)-2,5-diphenyltetrazoliumbromide
(MTT) (Sigma-Aldrich) assay.
Multicolor flow cytometry analysis
[0132] Tumour tissues were dissected into small pieces and digested with RPMI 1640 with
0.1mg/ml collagenase D and 50U/ml DNase I for 1h at 37°C on a shaking platform. The
cell suspension was filtered through a 70-µm cell strainer and centrifuged at 700
g for 10 min. The cell pellet was resuspended in PBS plus 1% bovine serum albumin
(BSA) for cell surface marker analysis. For intracellular cytokine staining, stimulation
was performed for the isolated cell suspension before surface marker staining. In
brief, the primary isolated cells were incubated in RPMI 1640 containing phorbol myristate
acetate (PMA), ionomycin, and monensin for 4 hours at 37°C. The stimulated cells were
then permeabilized by an intracellular staining kit (eBioscience) and stained with
anti-IFNγ, anti-TNF-α, and anti-GZMB overnight before multicolor flow cytometry analysis.
Bacteria whole genome sequencing (WGS)
[0133] The WGS of the 3 bacterial strains was conducted by Beijing Novogene Technology.
Bacterial genomic DNAs were extracted by using the ZR Faecal DNA MiniPrep (Zymo Research,
Irvine, CA) following the manufacturer's instructions.
Statistical analysis
[0134] Results are expressed as mean ± standard deviation (SD). ANOVA was used to compare
differences among multiple groups, and post-hoc analysis was performed by Tukey's
multiple comparisons test. P-values < 0.05 indicate statistical significance. Statistical
tests were performed in GraphPad Prism (V8.0, GraphPad Software Inc., San Diego, CA).
RESULTS
S. salivarius (K12) and its conditioned medium inhibit the viability of colon cancer cells
[0135] To evaluate the strain-specific anti-CRC effect of
S. salivarius, colon cancer cells, including HCT116, Lovo, and colon normal epithelial cell line
NCM460 were cocultured with 3 different strains of
S.
salivarius (MOI = 200, 2h).
E. coli strain MG1655 was used as a control. We found that, compared with
S. salivarius (M18) and
S. salivarius (Human), only
S. salivarius (K12) suppressed the CRC cells' viability (
Figure 21A). Concordantly, the conditioned medium from
S. salivarius (K12) (K12.CM) also revealed a solely inhibitory effect in suppressing the viability
of colon cancer cells, but not the normal epithelial cells (
Figure 21B), indicating that only the
S. salivarius (K12) strain contribute to the direct antitumourigenic effect.
[0136] We next determined whether such a strain-specific function was attributed to the
specific genetic background, 3
rd generation sequencing was then performed on the 3 different strains of
S.
salivarius. We discovered that
S. salivarius (K12) contains a gene cluster responsible for EPS production (
Figure 21C). Those results indicate that
S. salivarius containing EPS gene cluster can suppress the viability of CRC cells.
S. salivarius (K12) protects against intestinal tumourigenesis and augments effector CD8+ T-cell
infiltration in Apcmin/+ mice
[0137] To investigate the effect of
S. salivarius (K12) on colorectal tumourigenesis, the inventors gavaged the mice with
S. salivarius (K12), BHI and a non-tumourigenic
E. coli strain MG1655 were used as plain culture broth and bacteria control, respectively
(
Figure 22A). After 10-week gavage, they observed that administration of
S. salivarius (K12) significantly reduced the tumour number (
Figure 22B) and tumour size (
Figure 22C) in the colon and small intestine of
Apcmin/+ mice. They also examined the change of immune landscape induced by
S.
salivarius (K12). It was found that tumour-infiltrating cytotoxic T cells (CD8+) were significantly
enhanced in the
S. salivarius (K12)-treated
Apcmin/+ mice as compared with the BHI groups. The CD8+ effector T cells, granzyme B (GZMB)+
CD8+ T cells also enhanced significantly in the
S. salivarius (K12)-treated
Apcmin/+ mice (
Figure 22D), indicating the
S.
salivarius (K12) can abrogate intestinal tumourigenesis and modulate immune microenvironment
in
Apcmin/+ mice.
S. salivarius (K12) protects against intestinal tumourigenesis and augments effector CD8+ T-cell
infiltration in AOM/DSS-induced CRC mice
[0138] To validate the tumour-suppressive and immune modulatory effect of
S. salivarius (K12) on colorectal tumourigenesis, the inventors established a colitis-associated
CRC mouse model, in which DSS-promoted, AOM-induced CRC mice were treated with
S. salivarius (K12). BHI and a non-tumourigenic
E. coli strain MG1655 were used as plain culture broth and bacteria control, respectively.
F. prausnitzii, a CRC immune-modulating bacteria, was used as a positive control (
Figure 23A). After 14-week gavage, colonoscopy identified that colon tumour sizes in
S. salivarius (K12) treated group were visually smaller than the
E. coli MG1655 or broth control groups (
Figure 23B). In accordance with this observation, the inventors observed consistent results
as in the
Apcmin/+ mice (
Figure 23C and Figure 23D).
S. salivarius (K12) protects against intestinal tumourigenesis and augments effector CD8+ T-cell
infiltration in MC38 syngeneic mouse model
[0139] MC38 syngeneic model is an immunoresponsive murine tumour model. The inventors administered
S. salivarius (K12) to the MC38 (MSI-H) syngeneic mouse model. BHI was used as broth control,
F. prausnitzii was used as a positive control (
Figure 24A).
S.
salivarius (K12) significantly alleviate tumour growth and tumour weight (
Figure 24B). Of note, the suppressive effect was similar to
F.
prausnitzii (
Figure 24C). Moreover, the
S.
salivarius (K12) monotherapy also enhanced the tumour-infiltrating effector CD8+ cytotoxic T
cells (
Figure 24D). These results indicate that
S.
salivarius (K12) also functions in the MC38 syngeneic model to strengthen antitumour immunity.
S. salivarius (K12) reprograms the tumour immune microenvironment to synergize with anti-PD1 to
suppress colorectal tumourigenesis in the CT26 syngeneic model
[0140] CT26 (MSS CRC cell line) syngeneic model is an immunotherapy resistance murine tumour
model. No single-agent anti-PD1/PD-L1 therapy has shown any efficacy against this
tumour. To investigate whether
S. salivarius (K12) plus anti-PD1 exerts a synergistic CRC-suppressive effect, the CT26 syngeneic
model was treated with
S. salivarius (K12). BHI and
E.coli were used as control. After 2 weeks of pre-treatment, the CT 26 mouse CRC cells were
transplanted to the flank of the mice. When the tumour developed, anti-PD1 antibody
or IgG isotype control was intraperitoneally injected into the mice (
Figure 25A and Figure 25B). Monotherapy of
S. salivarius (K12) slightly decreased the tumour size and tumour weight (
Figure 25C). Most strikingly, combined
S. salivarius (K12) with anti-PD1 led to an obvious reduction of the tumour size and tumour weight
(Figure 25C) as compared with
S. salivarius (K12) alone or
BHI/
E. coli + anti-PD 1, indicating combined
S. salivarius (K12) with anti-PD synergistically abrogates CT26 syngeneic model. The inventors
also evaluated the tumour-infiltrating CD8+ cytotoxic T cells, they found the combination
therapy led to the significant induction of effector CD8+ T cells
(Figure 25D). Collectively, these results indicate that
S. salivarius (K12) enhances antitumour immunity in CRC.
DISCUSSION
[0141] In this study, the inventors demonstrate for the first time the strain-specific anti-CRC
effect of
S. salivarius. Oral administration of the
S. salivarius K12, which contains the EPS production gene cluster suppresses tumour formation in
3 murine models of CRC (
Apcmin/+, AOM/DSS, and MC38 syngeneic models). The tumour-infiltrating effector CD8+ T cells
were also enhanced in the
S. salivarius K12-treated CRC mice. Furthermore,
S. salivarius K12 synergistically with anti-PD1 in the immunotherapy resistance murine tumour model
(CT26 syngeneic model) to suppress the tumour size and enhance the effector CD8+ T
cells infiltration. These results strongly illustrate
S. salivarius K12 as a prophylactic for modulating antitumour immunity and improving anti-PD1 response.
[0142] All patents, patent applications, and other publications, including GenBank Accession
Numbers and equivalents, cited in this application are incorporated by reference in
the entirety for all purposes.